Cmp-dependent Sialidase Activity

SOBEK; Harald ;   et al.

Patent Application Summary

U.S. patent application number 17/355962 was filed with the patent office on 2021-11-04 for cmp-dependent sialidase activity. The applicant listed for this patent is Roche Diagnostics Operations, Inc.. Invention is credited to Michael GREIF, Sebastian MALIK, Harald SOBEK, Marco THOMANN.

Application Number20210340585 17/355962
Document ID /
Family ID1000005725356
Filed Date2021-11-04

United States Patent Application 20210340585
Kind Code A1
SOBEK; Harald ;   et al. November 4, 2021

CMP-DEPENDENT SIALIDASE ACTIVITY

Abstract

The present disclosure is directed to the properties of certain glycosyltransferase variants having N-terminal truncation deletions or internal deletions. Any of the mutants disclosed in here exhibit .alpha.-2,6-sialyltransferase enzymatic activity in the presence of CMP-activated sialic acid as co-substrate, and in the presence of a suitable acceptor site. A fundamental finding documented in the present disclosure is that such enzyme are not only capable of catalyzing transfer of a sialidyl moiety but they are also capable of catalyzing hydrolytic cleavage of terminally bound sialic acid from a glycan.


Inventors: SOBEK; Harald; (Biberach, DE) ; GREIF; Michael; (Penzberg, DE) ; THOMANN; Marco; (Penzberg, DE) ; MALIK; Sebastian; (Antdorf, DE)
Applicant:
Name City State Country Type

Roche Diagnostics Operations, Inc.

Indianapolis

IN

US
Family ID: 1000005725356
Appl. No.: 17/355962
Filed: June 23, 2021

Related U.S. Patent Documents

Application Number Filing Date Patent Number
15629040 Jun 21, 2017 11078511
17355962
PCT/EP2015/080741 Dec 21, 2015
15629040

Current U.S. Class: 1/1
Current CPC Class: C12N 9/1081 20130101; C12Y 301/03 20130101; C12Y 204/99001 20130101; C07K 2317/41 20130101; C12P 21/005 20130101; C07K 16/00 20130101; C12P 19/44 20130101
International Class: C12P 21/00 20060101 C12P021/00; C12N 9/10 20060101 C12N009/10; C12P 19/44 20060101 C12P019/44; C07K 16/00 20060101 C07K016/00

Foreign Application Data

Date Code Application Number
Dec 22, 2014 EP 14199648.8

Claims



1-10. (canceled)

11. A method of producing in vitro a sialylated target molecule, the method comprising the steps of a) providing an aqueous composition according to claim 1; (b) forming one or more terminal antennal N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine residue(s) by incubating the aqueous composition of step (a), thereby reacting cytidine-5'-monophospho-N-acetylneuraminic acid as co-substrate, thereby forming 5'-cytidine-monophosphate; and (c) hydrolyzing the phosphoester bond of the 5'-cytidine-monophosphate formed in step (b), thereby reducing 5'-cytidine-monophosphate-mediated inhibition, thereby maintaining the activity of the soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I; thereby producing in vitro the sialylated target molecule.

12. The method according to claim 11, wherein the method is performed at a temperature of 0.degree. C. to 40.degree. C.

13. The method according to claim 11, wherein steps (b) and (c) are performed in the same vessel.

14. The method according to claim 11, wherein steps (b) and (c) are performed for a period selected from the group consisting of 2 h to 96 h, 2 h to 23 h, 2 h to 6 h, and about 2 h.

15. The method according to claim 11, wherein steps (b) and (c) are performed for a period selected from the group consisting of 6 h to 96 h, 6 h to 23 h, and about 6 h.

16. The method according to claim 11, wherein steps (b) and (c) are performed for a period selected from the group consisting of 23 h to 96 h, and about 23 h.

17. The method according to claim 11, wherein steps (b) and (c) are performed for a period of about 96 h.
Description



CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application is a divisional application of U.S. application Ser. No. 15/629,040 (published as U.S. Publication No. 2017/0298405) filed Jun. 21, 2017, which is a continuation application of International Application No. PCT/EP2015/080741 filed Dec. 21, 2015, claiming priority to European Application No. 14199648.8 filed Dec. 22, 2014, the disclosures of which are hereby incorporated by reference in their entirety.

BACKGROUND

[0002] The present disclosure is directed to the properties of certain glycosyltransferase variants having N-terminal truncation deletions or internal deletions. Any of the mutants disclosed in here exhibit .alpha.-2,6-sialyltransferase enzymatic activity in the presence of CMP-activated sialic acid as co-substrate, and in the presence of a suitable acceptor site. A fundamental finding documented in the present disclosure is that such enzymes are not only capable of catalyzing transfer of a sialidyl moiety but they are also capable of catalyzing hydrolytic cleavage of terminally bound sialic acid from a glycan. Particularly it was found that in the presence of cytidine-5'-monophosphate (CMP) glycosyltransferase activity is inhibited, and sialidase activity is stimulated. Sialidase activity was found to be dependent on the presence of a particular stretch of amino acids (position 90 to 108) in the polypeptide sequence of the reference (wildtype) hST6Gal-I polypeptide. Deletion of this sequence portion in an N-terminal truncation variant was found to abolish sialidase activity, notably both in the presence and in the absence of CMP. Thus, disclosed are compositions, uses and methods employing the CMP-mediated feed-back regulation documented herein.

[0003] Contrary to previous findings .alpha.-2,6-sialyltransferase mutants were found to exhibit sialidase enzymatic activity, particularly (but not limited to) a variant of human .beta.-galactoside-.alpha.-2,6-sialyltransferase I (hST6Gal-I; wildtype amino acid sequence see SEQ ID NO:1) with a truncation deletion involving the first 89 N-terminal amino acids of the respective wild-type polypeptide (i.e. a mutant with the amino acid sequence of SEQ ID NO:2). A fundamental finding documented in the present disclosure is that this mutant enzyme is not only capable of catalyzing transfer of a sialidyl moiety; in fact, the .alpha.-2,6-sialyltransferase variant is also capable of catalyzing hydrolytic cleavage of terminally bound sialic acid from a glycan. The present disclosure further reports the unexpected observation of feed-back inhibition. Particularly it was found that in the presence of cytidine-5'-monophosphate (CMP) glycosyltransferase activity is inhibited, and sialidase activity is stimulated. Even more surprising, not only the deletion mutant involving the first 89 N-terminal amino acids but also other N-terminal truncation variants of human .beta.-galactoside-.alpha.-2,6-sialyltransferase I (hST6Gal-I) were found to exhibit sialidase enzymatic activity. However, sialidase activity was found to be dependent on the presence of a particular stretch of amino acids (position 90 to 108, see FIG. 1) in the polypeptide sequence of the reference (wildtype) hST6Gal-I polypeptide according to SEQ ID NO:1. Deletion of this sequence portion in an N-terminal truncation variant was found to abolish sialidase activity, notably both in the presence and in the absence of CMP. Thus, disclosed are compositions, uses and methods employing the CMP-mediated feed-back regulation documented herein, particularly directed to controlled hydrolysis of the .alpha.2,6 glycosidic bond in a N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine moiety. Further, compositions, uses and methods with a CMP-insensitive hST6Gal-I are disclosed.

[0004] Transferases (EC 2) catalyze transfer of a functional group from one substance to another. Glycosyltransferases, a superfamily of enzymes, are involved in synthesizing the carbohydrate portions of glycoproteins, glycolipids and glycosaminoglycans. Specific glycosyltransferases synthesize oligosaccharides by the sequential transfer of the monosaccharide moiety of an activated sugar donor to an acceptor molecule. Hence, a "glycosyltransferase" catalyzes the transfer of a sugar moiety from its nucleotide donor to an acceptor moiety of a polypeptide, lipid, glycoprotein or glycolipid. This process is also known as "glycosylation". A carbohydrate portion which is structural part of e.g. a glycoprotein is also referred to as "glycan". Glycans constitute the most prevalent of all known post-translational protein modifications. Glycans are involved in a wide array of biological recognition processes as diverse as adhesion, immune response, neural cell migration and axonal extension. As structural part of glycoproteins glycans also have a role in protein folding and the support of protein stability and biological activity.

[0005] In glycosyltransferase catalysis, the monosaccharide units glucose (Glc), galactose (Gal), N-acetylglucosamine (GlcNAc), N-acetylgalactosamine (GalNAc), glucuronic acid (GlcUA), galacturonic acid (GalUA) and xylose are activated as uridine diphosphate (UDP)-.alpha.-D derivatives; arabinose is activated as a UDP-.beta.-L derivative; mannose (Man) and fucose are activated as GDP-.alpha.-D and GDP-.beta.-L derivatives, respectively; and sialic acid (=.beta.-D-Neu5Ac; =Neu5Ac; =SA; =NANA) is activated as a CMP derivative of sialic acid. CMP-activated sialic acid (=CMP-.beta.-D-Neu5Ac, see below) appears to be the only naturally occurring nucleotide sugar in the form of a nucleotide monophosphate.

[0006] Many different glycosyltransferases contribute to the synthesis of glycans. The structural diversity of carbohydrate portions of glycoproteins is particularly large and is determined by complex biosynthetic pathways. In eukaryotes the post-translational biosynthesis of the glycan-part of glycoproteins takes place in the lumen of the endoplasmatic reticulum ("ER") and the Golgi apparatus. A single (branched or linear) carbohydrate chain of a glycoprotein is typically a N- or an O-linked glycan. During post-translational processing, carbohydrates are typically connected to the polypeptide via asparagine ("N-linked glycosylation"), or via serine or threonine ("O-linked glycosylation"). Synthesis of a glycan, no matter whether N- or O-linked (="N-/O-linked") is effected by the activity of several different membrane-anchored glycosyltransferases. A glycoprotein may comprise one or more glycan-connected amino acids (="glycosylation sites"). A specific glycan structure may be linear or branched. Branching is a notable feature of carbohydrates which is in contrast to the linear nature typical for DNA, RNA, and polypeptides. Combined with the large heterogeneity of their basic building blocks, the monosaccharides, glycan structures exhibit high diversity. Furthermore, in members of a particular glycoprotein species the structure of a glycan attached to a particular glycosylation site may vary, thus resulting in microheterogeneity of the respective glycoprotein species, i.e. in a species sharing the same amino acid sequence of the polypeptide portion.

[0007] A sialyltransferase (="ST") is a glycosyltransferase that catalyzes transfer of a sialic acid residue from a donor compound to (i) a terminal monosaccharide acceptor group of a glycolipid or a ganglioside, or (ii) to a terminal monosaccharide acceptor group of an N-/O-linked glycan of a glycoprotein. For the purpose of the present disclosure, the donor compound is also referred to as "co-substrate". For mammalian sialyltransferases including human ST species there is a common donor compound which is cytidine-5'-monophospho-N-acetylneuraminic acid (=CMP-.beta.-D-Neu5Ac=CMP-Neu5Ac=CMP-NANA; =CMP-sialic acid; =CMP-SA). Well known to the skilled person, CMP-sialic acid is a specific embodiment of a donor compound for a sialyltransferase; further, there exist functional equivalents including but not limited to CMP-9-fluoresceinyl-sialic acid. Transfer and covalent coupling of a sialic acid residue (or the functional equivalent thereof) to a receptor site is also referred to as "sialylating" and "sialylation".

[0008] In the glycan structure of a sialylated glycoprotein the (one or more) sialyl moiety (moieties) is (are) usually found in the terminal position of an oligosaccharide. Thus, depending on the amount of sialylated sites, one or more sialic acid residue(s) can form part of a glycan moiety of a given glycoprotein. Owing to the terminal, i.e. exposed position, sialic acid can participate in many different biological recognition phenomena and serve in different kinds of biological interactions. In a glycoprotein more than one sialylation site may be present, i.e. a site capable of serving as a substrate for a sialyltransferase and being an acceptor group suitable for the transfer of a sialic acid residue. Such more than one site can in principle be the termini of a plurality of linear glycan portions anchored at different glycosylation sites of the glycoprotein. Additionally, a branched glycan may have a plurality of sites where sialylation can occur.

[0009] According to current knowledge, a terminal sialic acid residue can be found (i) .alpha.2.fwdarw.3 (.alpha.2,3) linked to galactosyl-R, (ii) .alpha.2.fwdarw.6 (.alpha.2,6) linked to galactosyl-R, (iii) .alpha.2.fwdarw.6 (.alpha.2,6) linked to N-acetylgalactosaminidyl-R, (iv) .alpha.2.fwdarw.6 (.alpha.2,6) linked to N-acetylglucosaminidyl-R, and (v) .alpha.2.fwdarw.8/9 (.alpha.2,8/9) linked to sialidyl-R, wherein --R denotes the rest of the acceptor substrate moiety. Hence, a sialyltransferase active in the biosynthesis of sialylconjugates is generally named and classified according to its respective monosaccharide acceptor substrate and according to the 3, 6 or 8/9 position of the glycosidic bond it catalyzes. Accordingly, in the literature known to the art, e.g. in Patel R Y, et al, Glycobiology 16 (2006) 108-116, reference to eukaryotic sialyltransferases is made such as (i) ST3Gal, (ii) ST6Gal, (iii) ST6GalNAc, or (v) ST8Sia, depending on the hydroxyl position of the acceptor sugar residue to which the Neu5Ac residue is transferred while forming a glycosidic bond. Reference to sialyltransferases in a more generic way can also be made e.g. as ST3, ST6, ST8; thus, "ST6" specifically encompasses the sialyltransferases catalyzing an .alpha.2,6 sialylation.

[0010] The disaccharide moiety .beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-glucosamine (=Gal.beta.1,4GlcNAc) is a frequent terminal residue of the antennae of N-linked glycans of glycoproteins, but may be also present in O-linked glycans and in glycolipids. In addition, a terminal Gal.beta.1,4GlcNAc moiety can be generated in certain target glycoproteins as a result of galactosyltransferase enzymatic activity. The enzyme .beta.-galactoside-.alpha.2,6-sialyltransferase (="ST6Gal") is able to catalyze .alpha.2,6-sialylation of a terminal Gal.beta.1,4GlcNAc acceptor moiety of a glycan or a branch of a glycan, also known to the art as "antenna". For general aspects thereof, reference is made to the document of DallOlio F. Glycoconjugate Journal 17 (2000) 669-676. In human and in other mammals there appear to be several species (isozymes) of ST6Gal. The present disclosure particularly discloses human .beta.-galactoside-.alpha.-2,6-sialyltransferase I (=hST6Gal-I; EC 2.4.99.1 according to IUBMB Enzyme Nomenclature) and variants thereof, but is not limited thereto.

[0011] The ST6 group of sialyltransferases comprises two subgroups, ST6Gal and ST6GalNAc. The activity of ST6Gal enzymes catalyzes transfer of a Neu5Ac residue to the C6 hydroxyl group of a free galactosyl residue being part of terminal Gal.beta.1,4GlcNAc in a glycan or an antenna of a glycan, thereby forming in the glycan a terminal sialic acid residue .alpha.2.fwdarw.6 linked to the galactosyl residue of the Gal.beta.1,4GlcNAc moiety. The resulting newly formed terminal moiety in the glycan is Neu5Ac.alpha.2,6Gal.beta.1,4GlcNAc.

[0012] The wild-type polypeptide of human .beta.-galactoside-.alpha.-2,6-sialyltransferase I (hST6Gal-I) at the time of filing of the present document was disclosed as "UniProtKB/Swiss-Prot: P15907.1" in the publically accessible NCBI database (http://www.ncbi.nlm.nih.gov/protein/115445). Further information including coding sequences are provided as hyperlinks compiled within the database entry "Gene ID: 6480" (http://www.ncbi.nlm.nih.gov/gene/6480).

[0013] Mammalian sialyltransferases share with other mammalian Golgi-resident glycosyltransferases a so-called "type II architecture" with (i) a short cytoplasmic N-terminal tail, (ii) a transmembrane fragment followed by (iii) a stem region of variable length and (iv) a C-terminal catalytic domain facing the lumen of the Golgi apparatus (Donadio S. et al. in Biochimie 85 (2003) 311-321). Accordingly, a "soluble" sialyltransferase with an N-terminal truncation deletion lacks at least the elements (i) and (ii) of the type II architecture. Mammalian sialyltransferases appear to display significant sequence homology in their catalytic domain. Recent data regarding structure and function of hST6Gal-I are disclosed in Kuhn B. et al. (Biol. Crystallography D69 (2013) 1826-1838).

[0014] In certain mammals including mouse, rat and humans, ST6Gal has widespread tissue distribution. It is particularly abundant in liver, the major site of serum glycoprotein synthesis (Weinstein J. et al. J. Biol. Chem 257 (1982) 13835-13844). On the one hand sialyltransferase exists predominantly in a membrane-bound form within the Golgi and trans-Golgi network where it participates in the posttranslational modification of newly synthesized secretory or cell surface glycoproteins. On the other hand, a soluble form of ST6Gal-I exists in the serum (Kim Y. S. et al Biochim. Biophys Acta 244 (1971) 505-512; Dalziel M. et al. Glycobiology 9 (1999) 1003-1008) and predominantly is derived from the liver (Kaplan, H. A. et al. J. Biol. Chern. 258 (1983) 11505-11509; van Dijk, W. et al. Biochem. Cell. Biol. 64 (1986) 79-84; Dalziel M. et al. (supra)) by a proteolytic event that liberates the catalytic domain from its membrane anchor (Kaplan et al., supra; Weinstein, J. et al. J. Biol. Chem. 262 (1987) 17735-17743). Thus, any variant of an originally membrane-anchored glycosyltransferase with an N-terminal truncation comprising the membrane anchor is encompassed by the term "soluble variant". Soluble variants can exemplarily be generated by proteolytically removing the portion with the membrane anchor from the protein, or by expressing a variant nucleic acid sequence encoding a N-terminally truncated form of the original protein wherein the truncation includes the membrane anchor (transmembrane fragment, element (ii) of the type II architecture).

[0015] Donadio S. et al. (supra) recombinantly expressed several N-terminally truncated variants of hST6Gal-I without the membrane anchor in CHO cells. The authors found that N-terminal deletions comprising the first 35, 48, 60 and 89 amino acids yielded variants of hST6Gal-I which were enzymatically active and capable of transferring sialic acid to exogenous acceptors.

[0016] Glycosylation is an important posttranslational modification of proteins influencing protein folding, stability and regulation of the biological activity. The sialyl residue is usually exposed at the terminal position of an N-glycan and therefore, a major contributor to biological recognition and ligand function. As an important example, IgG with glycans featuring terminal sialic acid residues were found to induce reduced inflammatory response and showed an increase in serum half-life. Therefore, use of glycosyltransferases for enzymatic synthesis of defined glycan structures is becoming an engineering tool towards direct in vitro N-glycosylation of therapeutic proteins, and particularly therapeutic monoclonal antibodies.

[0017] Since glycosyltransferases of prokaryotic origin usually do not act on complex glycoprotein structures, sialyltransferases of mammalian origin are preferred for in vitro glycoengineering purposes. For example, Barb et al. (2009) prepared highly sialylated forms of the Fc fragment of immunoglobulin G using isolated human ST6Gal-I. However, the access to recombinant hST6Gal-I for such applications is still limited due to low expression yield and/or poor activity of hST6Gal-I recombinantly expressed in various hosts (methylotrophic yeast Pichia pastoris, cultured Spodoptera frugiperda cells, E. coli-based expression systems).

[0018] Kleineidam R. G. et al. Glycoconjugate Journal (1997) 14: 57-66 disclose a number of inhibitors of .alpha.-2,6-sialyltransferase from rat liver. Specifically, 70%, 40%, 39% and 71% inhibition of .alpha.-2,6-sialyltransferase was observed in the presence of Cytidine, 2'-CMP, 3'-CMP and 5'-CMP, respectively, wherein each inhibitor was tested at a concentration of 0.25 mM.

[0019] While the use of mammalian glycosyltransferases for in vitro sialylating a glycosylated target molecule such as a glycoprotein or a glycolipid is known to the art, the opposite reaction (sialidase activity, hydrolytic cleavage of a terminal sialyl residue from a glycan moiety) is typically provided by a neuraminidase, so far. The original finding by the present inventors is, however, that a soluble variant of a sialyltransferase of mammalian origin displays sialidase activity in the presence of CMP. In fact, the specific example of a soluble variant of human .beta.-galactoside-.alpha.-2,6-sialyltransferase I lacking the transmembrane domain by means of a N-terminal truncation can be used for both, (i) sialylation of a target glycoprotein and (ii) hydrolytic cleavage of sialyl residues from a sialylated target glycoprotein. Depending on the presence of CMP and the interaction of CMP with the soluble variant, sialylation can be controlled quantitatively. In specific embodiments involving target molecules with two or more antennal glycan acceptor sites, the present disclosure provides means, methods and conditions allowing to sialylate just one out of the several acceptor sites, as well as sialylating two or more, or even all acceptor sites of the target molecule.

[0020] This paves the way for a number of different approaches, particularly in the field of in vitro glycoengineering of immunoglobulins, and also of other glycosylated target molecules.

SUMMARY

[0021] In a first aspect and a specific embodiment of all other aspects as disclosed herein there is disclosed an aqueous composition comprising [0022] (a) a soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprising the amino acid motif from position 90 to position 108 in SEQ ID NO:1; [0023] (b) cytidine-5'-monophospho-N-acetylneuraminic acid, or a functional equivalent thereof; [0024] (c) a glycosylated target molecule selected from a glycoprotein and a glycolipid, the target molecule comprising one or more antenna(e), at least one antenna having as a terminal structure a .beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-glucosamine moiety with a hydroxyl group at the C6 position in the galactosyl residue; [0025] (d) an aqueous solution permitting glycosyltransferase enzymatic activity; wherein the aqueous composition further comprises an enzyme capable of hydrolyzing the phosphoester bond in 5'-cytidine-monophosphate under conditions permitting glycosyltransferase enzymatic activity.

[0026] In a second aspect and a specific embodiment of all other aspects as disclosed herein there is disclosed the use of an enzyme capable of the hydrolyzing phosphoester bond in 5'-cytidine-monophosphate for maintaining sialyltransferase enzymatic activity and/or inhibiting sialidase enzymatic activity in a composition according to the first aspect.

[0027] In a third aspect and a specific embodiment of all other aspects as disclosed herein there is disclosed a method of producing in vitro a sialylated target molecule, the method comprising the steps of [0028] (a) providing an aqueous composition according to any of the claims 1 and 2; [0029] (b) forming one or more terminal antennal N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine residue(s) by incubating the aqueous composition of step (a), thereby reacting cytidine-5'-monophospho-N-acetylneuraminic acid, or a functional equivalent thereof, as co-substrate, thereby forming 5'-cytidine-monophosphate; [0030] (c) hydrolyzing the phosphoester bond of the 5'-cytidine-monophosphate formed in step (b), thereby reducing 5'-cytidine-monophosphate-mediated inhibition, thereby maintaining the activity of the soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I; thereby producing in vitro the sialylated target molecule.

[0031] In a fourth aspect and a specific embodiment of all other aspects as disclosed herein there is disclosed a preparation of sialylated immunoglobulins, each immunoglobulin having a plurality of acceptor sites for human .beta.-galactoside-.alpha.-2,6-sialyltransferase I, wherein less than about 25% of the acceptor sites in the preparation of sialylated immunoglobulins are not sialylated, and about 75% or more are sialylated, wherein the preparation is obtained by a method according to the third aspect.

[0032] In a fifth aspect and a specific embodiment of all other aspects as disclosed herein there is disclosed the use of a soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprising the amino acid motif from position 90 to position 108 in SEQ ID NO:1 for in vitro hydrolyzing in the presence of 5'-cytidine-monophosphate the .alpha.2,6 glycosidic bond in a N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine moiety, the moiety being a terminal structure of a glycan in a sialylated glycoprotein or glycolipid.

DESCRIPTION OF THE FIGURES

[0033] FIG. 1 Representation of the amino acid sequence of the wild-type hST6Gal-I polypeptide (SEQ ID NO:1), and the N-terminal portions thereof which are truncated in the deletion variants as disclosed herein. The deleted positions in the truncations are symbolized by "X". Underlined are the amino acid at positions 90-108 (SEQ ID NO:1) found to be essential for CMP-induced Sialidase activity.

[0034] FIG. 2 SDS-PAGE gel after electrophoresis and staining of the .DELTA.89 hST6Gal-I variant transiently expressed in and secreted from HEK cells. Lane 1 shows a size-standard, molecular weights in kDa according to the standard are indicated to the left. Lane 2: Purified .DELTA.89 hST6Gal-I truncation variant (5 .mu.g of protein were loaded on the gel).

[0035] FIG. 3 SDS gel after electrophoresis and staining of the .DELTA.108 hST6Gal-I variant transiently expressed in and secreted from HEK cells. Lane 1 shows a size-standard, molecular weights in kDa according to the standard are indicated to the left. Lane 2: .DELTA.108 hST6Gal-I truncation variant (5 .mu.g of protein were loaded on the gel).

[0036] FIG. 4 Time course of sialylation of IgG4 MAB using recombinant .DELTA.89 hST6Gal-I.

[0037] FIG. 5 Kinetics of formation of G2+2SA and G2+1SA, catalyzed by recombinant .DELTA.89 hST6Gal-I, as shown by mass spectra taken as a basis for determination of the relative content of the different sialylated target molecule species.

[0038] FIG. 6 Inhibition of sialidase activity of recombinant .DELTA.89 hST6Gal-I by CTP. The relative content of antibodies with glycan with terminal galactose residues (G2+0SA, "asialo"), mono-sialylated glycan (G2+1SA) and bi-sialylated glycan (G2+2SA) is shown for different time points.

[0039] FIG. 7 CMP-dependent sialidase activity of .DELTA.89 hST6Gal-I: Incubation of purified IgG1 MAB G2+2SA with .DELTA.89 hST6Gal-I in the absence of CMP. The relative content of antibodies with glycans with terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) are shown for different time points.

[0040] FIG. 8 CMP-dependent sialidase activity of .DELTA.89 hST6Gal-I: Incubation of purified IgG1 MAB G2+2SA with .DELTA.89 hST6Gal-I in the presence of CMP. The relative content of antibodies with glycans with terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) are shown for different time points.

[0041] FIG. 9 CMP-dependent sialidase activity of .DELTA.89 hST6Gal-I: Incubation of purified IgG1 MAB G2+2SA with .DELTA.108 hST6Gal-I in the presence of CMP. The relative content of antibodies with glycans with terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) are shown for different time points.

[0042] FIG. 10 CMP-dependent sialidase activity of .DELTA.89 hST6Gal-I: Incubation of purified IgG1 MAB G2+2SA with delta57ST3-Gal-I in the presence of CMP. The relative content of antibodies with glycans with terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) are shown for different time points.

[0043] FIG. 11 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of 5'-nucleotidase CD73 in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. The amount of 5'-nucleotidase used was 0-0.5 .mu.g. Negative control: 0 .mu.g 5'-nucleotidase CD73.

[0044] FIG. 12 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of 5'-nucleotidase CD73 in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. Sialylation reaction mixture with 0.1 .mu.g 5'-nucleotidase CD73.

[0045] FIG. 13 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of 5'-nucleotidase CD73 in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. Sialylation reaction mixture with 0.25 .mu.g 5'-nucleotidase CD73.

[0046] FIG. 14 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of 5'-nucleotidase CD73 in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. Sialylation reaction mixture with 0.5 .mu.g 5'-nucleotidase CD73.

[0047] FIG. 15 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of alkaline phosphatase in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. The amount of 5'-nucleotidase used was 0-100 .mu.g. Negative control: 0 .mu.g alkaline phosphatase.

[0048] FIG. 16 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of alkaline phosphatase in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. Sialylation reaction mixture with 1 .mu.g alkaline phosphatase.

[0049] FIG. 17 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of alkaline phosphatase in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. Sialylation reaction mixture with 5 .mu.g alkaline phosphatase.

[0050] FIG. 18 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of alkaline phosphatase in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. Sialylation reaction mixture with 10 .mu.g alkaline phosphatase.

[0051] FIG. 19 Sialylation of IgG4 MAB with .DELTA.89 hST6Gal-I in the absence or presence of alkaline phosphatase in the sialylation reaction mixture. The relative content of antibodies with glycans with just terminal galactose residues (G2+0SA), monosialylated glycan (G2+1SA) and disialylated glycan (G2+2SA) is shown. Sialylation reaction mixture with 100 .mu.g alkaline phosphatase.

DETAILED DESCRIPTION

[0052] The terms "a", "an" and "the" generally include plural referents, i.e. "one or more", unless the context clearly indicates otherwise. As used herein, "plurality" is understood to mean more than one. For example, a plurality refers to at least two, three, four, five, or more. Unless specifically stated or obvious from context, as used herein, the term "or" is understood to be inclusive.

[0053] Unless specifically stated or obvious from context, as used herein, the term "about" is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. About can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05%, or 0.01% of the stated value. Unless otherwise clear from context, all numerical values provided herein can be modified by the term "about".

[0054] The term "amino acid" generally refers to any monomer unit that can be incorporated into a peptide, polypeptide, or protein. As used herein, the term "amino acid" includes the following twenty natural or genetically encoded alpha-amino acids: alanine (Ala or A), arginine (Arg or R), asparagine (Asn or N), aspartic acid (Asp or D), cysteine (Cys or C), glutamine (Gln or Q), glutamic acid (Glu or E), glycine (Gly or G), histidine (His or H), isoleucine (Ile or I), leucine (Leu or L), lysine (Lys or K), methionine (Met or M), phenylalanine (Phe or F), proline (Pro or P), serine (Ser or S), threonine (Thr or T), tryptophan (Trp or W), tyrosine (Tyr or Y), and valine (Val or V). In cases where "X" residues are undefined, these should be defined as "any amino acid." The structures of these twenty natural amino acids are shown in, e.g., Stryer et al., Biochemistry, 5th ed., Freeman and Company (2002). Additional amino acids, such as selenocysteine and pyrrolysine, can also be genetically coded for (Stadtman (1996) "Selenocysteine," Annu Rev Biochem. 65:83-100 and Ibba et al. (2002) "Genetic code: introducing pyrrolysine," Curr Biol. 12(13):R464-R466). The term "amino acid" also includes unnatural amino acids, modified amino acids (e.g., having modified side chains and/or backbones), and amino acid analogs. See, e.g., Zhang et al. (2004) "Selective incorporation of 5-hydroxytryptophan into proteins in mammalian cells," Proc. Natl. Acad. Sci. U.S.A. 101(24):8882-8887, Anderson et al. (2004) "An expanded genetic code with a functional quadruplet codon" Proc. Natl. Acad. Sci. U.S.A. 101(20):7566-7571, Ikeda et al. (2003) "Synthesis of a novel histidine analogue and its efficient incorporation into a protein in vivo," Protein Eng. Des. Sel. 16(9):699-706, Chin et al. (2003) "An Expanded Eukaryotic Genetic Code," Science 301(5635):964-967, James et al. (2001) "Kinetic characterization of ribonuclease S mutants containing photoisomerizable phenylazophenylalanine residues," Protein Eng. Des. Sel. 14(12):983-991, Kohrer et al. (2001) "Import of amber and ochre suppressor tRNAs into mammalian cells: A general approach to site-specific insertion of amino acid analogues into proteins," Proc. Natl. Acad. Sci. U.S.A. 98(25):14310-14315, Bacher et al. (2001) "Selection and Characterization of Escherichia coli Variants Capable of Growth on an Otherwise Toxic Tryptophan Analogue," J. Bacteriol. 183(18):5414-5425, Hamano-Takaku et al. (2000) "A Mutant Escherichia coli Tyrosyl-tRNA Synthetase Utilizes the Unnatural Amino Acid Azatyrosine More Efficiently than Tyrosine," J. Biol. Chem. 275(51):40324-40328, and Budisa et al. (2001) "Proteins with {beta}-(thienopyrrolyl)alanines as alternative chromophores and pharmaceutically active amino acids," Protein Sci. 10(7):1281-1292. To further illustrate, an amino acid is typically an organic acid that includes a substituted or unsubstituted amino group, a substituted or unsubstituted carboxy group, and one or more side chains or groups, or analogs of any of these groups. Exemplary side chains include, e.g., thiol, seleno, sulfonyl, alkyl, aryl, acyl, keto, azido, hydroxyl, hydrazine, cyano, halo, hydrazide, alkenyl, alkynl, ether, borate, boronate, phospho, phosphono, phosphine, heterocyclic, enone, imine, aldehyde, ester, thioacid, hydroxylamine, or any combination of these groups. Other representative amino acids include, but are not limited to, amino acids comprising photoactivatable cross-linkers, metal binding amino acids, spin-labeled amino acids, fluorescent amino acids, metal-containing amino acids, amino acids with novel functional groups, amino acids that covalently or noncovalently interact with other molecules, photocaged and/or photoisomerizable amino acids, radioactive amino acids, amino acids comprising biotin or a biotin analog, glycosylated amino acids, other carbohydrate modified amino acids, amino acids comprising polyethylene glycol or polyether, heavy atom substituted amino acids, chemically cleavable and/or photocleavable amino acids, carbon-linked sugar-containing amino acids, redox-active amino acids, amino thioacid containing amino acids, and amino acids comprising one or more toxic moieties.

[0055] The term "protein" refers to a polypeptide chain (amino acid sequence) as a product of the ribosomal translation process, wherein the polypeptide chain has undergone posttranslational folding processes resulting in three-dimensional protein structure. The term "protein" also encompasses polypeptides with one or more posttranslational modifications such as (but not limited to) glycosylation, phosphorylation, acetylation and ubiquitination.

[0056] Any protein as disclosed herein, particularly recombinantly produced protein as disclosed herein, may in a specific embodiment comprise a "protein tag" which is a peptide sequence genetically grafted onto the recombinant protein. A protein tag may comprise a linker sequence with a specific protease cleavage site to facilitate removal of the tag by proteolysis. As a specific embodiment, an "affinity tag" is appended to a target protein so that the target can be purified from its crude biological source using an affinity technique. For example, the source can be a transformed host organism expressing the target protein or a culture supernatant into which the target protein was secreted by the transformed host organism. Specific embodiments of an affinity tag include chitin binding protein (CBP), maltose binding protein (MBP), and glutathione-S-transferase (GST). The poly(His) tag is a widely-used protein tag which facilitates binding to certain metal chelating matrices.

[0057] Each of the terms "chimeric protein", "fusion protein" or "fusion polypeptide" equally refers to a protein whose amino acid sequence represents a fusion product of subsequences of the amino acid sequences from at least two distinct proteins. A fusion protein typically is not produced by direct manipulation of amino acid sequences, but, rather, is expressed from a "chimeric" gene that encodes the chimeric amino acid sequence.

[0058] The term "recombinant" refers to an amino acid sequence or a nucleotide sequence that has been intentionally modified by recombinant methods. By the term "recombinant nucleic acid" herein is meant a nucleic acid, originally formed in vitro, in general, by the manipulation of a nucleic acid by endonucleases, in a form not normally found in nature. Thus an isolated, mutant DNA polymerase nucleic acid, in a linear form, or an expression vector formed in vitro by ligating DNA molecules that are not normally joined, are both considered recombinant for the purposes of this invention. It is understood that once a recombinant nucleic acid is made and reintroduced into a host cell, it will replicate non-recombinantly, i.e., using the in vivo cellular machinery of the host cell rather than in vitro manipulations; however, such nucleic acids, once produced recombinantly, although subsequently replicated non-recombinantly, are still considered recombinant for the purposes of the invention. A "recombinant protein" or "recombinantly produced protein" is a protein made using recombinant techniques, i.e., through the expression of a recombinant nucleic acid as depicted above.

[0059] The term "host cell" refers to both single-cellular prokaryote and eukaryote organisms (e.g., mammalian cells, insect cells, bacteria, yeast, and actinomycetes) and single cells from higher order plants or animals when being grown in cell culture.

[0060] The term "glycosylation" denotes the chemical reaction of covalently coupling a glycosyl residue to an acceptor group. One specific acceptor group is a hydroxyl group, e.g. a hydroxyl group of another sugar. "Sialylation" is a specific form of glycosylation wherein the acceptor group is reacted with a sialic acid (=N-acetylneuraminic acid) residue. Such a reaction is typically catalyzed by a sialyltransferase enzyme using cytidine-5'-monophospho-N-acetylneuraminic acid as donor compound or co-substrate.

[0061] "Sialylation" is a specific embodiment of a result of glycosyltransferase enzymatic activity (sialyltransferase enzymatic activity in the particular case), under conditions permitting the same.

[0062] Generally, the skilled person appreciates that the aqueous composition in which glycosyltransferase enzymatic activity can take place (=under conditions "permitting glycosyltransferase enzymatic activity") needs to be buffered using a buffer salt such as Tris, MES, phosphate, acetate, or another buffer salt specifically capable of buffering in the pH range of pH 6 to pH 8, more specifically in the range of pH 6 to pH 7, even more specifically capable of buffering a solution of about pH 6.5. The buffer may further contain a neutral salt such as but not limited to NaCl. Further, in particular embodiments the skilled person may consider adding to the aqueous buffer a salt comprising a divalent cation such as Mg.sup.2+ or Mn.sup.2+, e.g., but not limited to, MgCl.sub.2 and MnCl.sub.2. In additional specific embodiments, the aqueous composition permitting glycosyltransferase enzymatic activity may comprise an antioxidant and/or a surfactant. Conditions permitting glycosyltransferase enzymatic activity known to the art include ambient (room) temperature, but more generally temperatures in the range of 0.degree. C. to 40.degree. C., particularly 10.degree. C. to 30.degree. C., particularly about 20.degree. C. While the above described conditions provide general conditions permitting such enzymatic activity, glycosyltransferase activity further requires the presence of an activated sugar donor (such as specifically CMP-Neu5Ac) as a co-substrate, in addition. However, the term "permitting glycosyltransferase enzymatic activity" is understood as not necessarily including the presence of the co-substrate. Thus, the term "permitting glycosyltransferase enzymatic activity" herein also includes conditions permitting the hydrolysis (sialidase) activity of a mammalian glycosyltransferase subject of the present disclosure, particularly hydrolysis activity in the presence of 5'-cytidine-monophosphate (CMP).

[0063] The term "glycan" refers to a poly- or oligosaccharide, i.e. to a multimeric compound which upon acid hydrolysis yields a plurality of monosaccharides. A glycoprotein comprises one or more glycan moieties which are covalently coupled to side groups of the polypeptide chain, typically via asparagine or arginine ("N-linked glycosylation") or via serine or threonine ("0-linked glycosylation").

[0064] The use of glycosyltransferases for enzymatic synthesis of complex glycan structures is an attractive approach to obtain complex bioactive glycoproteins. E.g. Barb et al. Biochemistry 48 (2009) 9705-9707 prepared highly potent sialylated forms of the Fc fragment of immunoglobulin G using isolated human ST6Gal-I. However, growing interest in the therapeutic application of glycoproteins leads to an increasing demand of glycosyltransferases including sialyltransferases. Different strategies to increase or modify the sialylation of glycoproteins were described by Bork K. et al. J. Pharm. Sci. 98 (2009) 3499-3508. An attractive strategy is sialylation in vitro of recombinantly produced proteins (such as but not limited to immunoglobulins and growth factors), particularly therapeutic proteins. To this end, several research groups described expression of sialyltransferases in transformed organisms and purification of the recombinantly produced sialyltransferases. As glycosyltransferases of prokaryotic origin usually do not act on complex glycoproteins (e.g. antibodies), sialyltransferases from mammalian origin were studied with preference.

[0065] Particular glycoproteins subject to the disclosures and all aspects of the present document and the aspects and embodiments herein comprise without limitation cell surface glycoproteins and glycoproteins present in soluble form in serum ("serum glycoprotein"), the glycoproteins particularly being of mammalian origin. A "cell surface glycoprotein" is understood to be glycoprotein of which a portion is located on and bound to the surface of a membrane, by way of a membrane anchor portion of the surface glycoprotein's polypeptide chain, wherein the membrane is part of a biological cell. The term cell surface glycoprotein also encompasses isolated forms of the cell surface glycoprotein as well as soluble fragments thereof which are separated from the membrane anchor portion, e.g. by proteolytic cleavage or by recombinant production of such soluble fragments. A "serum glycoprotein" is understood as a glycoprotein being present in serum, i.e. a blood protein present in the non-cellular portion of whole blood, e.g. in the supernatant following sedimentation of cellular blood components. Without limitation, a specifically regarded and embodied serum glycoprotein is an immunoglobulin. Particular immunoglobulins mentioned in here belong to the IgG group (characterized by Gamma heavy chains), specifically any of four the IgG subgroups. For the disclosures, aspects and embodiments herein the term "serum glycoprotein also encompasses a monoclonal antibody; monoclonal antibodies artificially are well known to the art and can be produced e.g. by hybridoma cells or recombinantly using transformed host cells. A further serum specific glycoprotein is a carrier protein such as serum albumin, a fetuin, or another glycoprotein member of the superfamily of histidine-rich glycoproteins of which the fetuins are members. Further, without limitation, a specifically regarded and embodied serum glycoprotein regarding all disclosures, aspects and embodiments herein is a glycosylated protein signalling molecule. A particular molecule of this group is erythropoietin (EPO).

[0066] For in vitro engineering of glycoproteins glycosyltransferases can be used as an efficient tool (Weijers 2008). Glycosyltransferases of mammalian origin are compatible with glycoproteins as substrates whereas bacterial glycosyltransferases usually modify simpler substrates like oligosaccharides. For this reason synthetic changes in the glycan moieties of glycoproteins are advantageously made using mammalian glycosyltransferases as tools of choice. However, for a large scale application of glycosyltransferases in glycoengineering availability of suitable enzymes in large (i.e. industrial) quantities is required. The disclosure herein particularly provides proteins with (i) hST6Gal-I sialyltransferase activity and (ii) sialidase activity which can be used for quantitatively controlled in vitro sialylation of target glycoproteins with one or more accessible galactosyl substrate moiety/moieties.

[0067] Importantly, the amino acid motif in human .beta.-galactoside-.alpha.-2,6-sialyltransferase I from position 90 to position 108 in SEQ ID NO:1 is required for the enzyme to be capable of exhibiting sialidase activity. At the same time, this amino acid motif is required for the enzyme to interact with 5'-CMP. Very remarkably a truncation deletion mutant, a soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I variant lacking the contiguous N-terminal stretch of the amino acids from position 1 to position 108 does not exhibit sialidase activity, not even in the presence of CMP. Thus, it was concluded that the amino acid motif in human .beta.-galactoside-.alpha.-2,6-sialyltransferase I from position 90 to position 108 in SEQ ID NO:1 is essential for these properties to be present.

[0068] Suitable targets to treat with sialidase activity include on the one hand asialoglycoproteins, i.e. glycoproteins from which sialic acid residues have been removed by the action of sialidases. On the other hand, bi-sialylated glycoproteins may serve as substrate for sialidase activity. Very advantageously, asialo-, mono-sialylated and bi-sialylated immunoglobulins are specific substrates, particularly immunoglobulins of the IgG class.

[0069] While expressing wild-type hST6Gal-I in the methylotrophic yeast Pichia pastoris and having targeted the expressed polypeptide to the secretory pathway of the host organism, different truncated variants of recombinantly produced hST6Gal-I were observed. Generally, hST6Gal-I derived proteins were chromatographically purified and analyzed, particularly by means of mass spectrometry and by way of determining the amino acid sequence from the N-terminus (Edman degradation). By these means truncations, particularly N-terminal truncations of hST6Gal-I were characterized in detail.

[0070] Several remarkable truncation variants were identified in the supernatants of transformed Pichia strains. The variants could possibly result from site-specific proteolytic cleavage during the course of secretion from the yeast cells, or result from endoproteolytic cleavage by one or more extracellular protease(s) present in the supernatant of cultured Pichia strains.

[0071] Each identified truncation variant was given a "delta" (=".DELTA.") designation indicating the number of the last amino acid position of the respective truncation deletion, counted from the N-Terminus of the wild-type hST6Gal-I polypeptide according to SEQ ID NO:1 The particular N-terminal truncation variants .DELTA.89 and .DELTA.108 of hST6Gal-I were recombinantly expressed and studied in more detail.

[0072] Expression vectors were constructed for expression of hST6Gal-I wild-type protein as well as of the .DELTA.89 and .DELTA.108 truncation variants in various host organisms including prokaryotes such as E. coli and Bacillus sp., yeasts such as Saccharomyces cerevisiae and Pichia pastoris, and mammalian cells such as CHO cells and HEK cells. Vectors with expression constructs for the .DELTA.89 and .DELTA.108 truncation variants of hST6Gal-I were assembled molecularly thereby providing the means of recombinantly producing the .DELTA.89 variant of human ST6Gal-I in several transformed host organisms. To facilitate purification of recombinantly expressed enzymes, the encoded truncation polypeptides encoded by the constructs optionally included a N-terminal His-tag, in specific embodiments.

[0073] An aspect and a specific embodiment of all other aspects as disclosed herein is a variant mammalian glycosyltransferase capable of catalyzing hydrolysis of the .alpha.2,6 glycosidic bond of a N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine moiety of a glycan in a glycoprotein. Particularly, the variant mammalian glycosyltransferase is capable of catalyzing formation of the .alpha.2,6 glycosidic bond of a N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine moiety in a glycoprotein glycan, thereby generating free N-acetylneuraminic acid. In a specific embodiment of all aspects as disclosed herein, the variant mammalian glycosyltransferase is a soluble enzyme comprising the amino acid motif from position 90 to position 108 in SEQ ID NO:1 disclosing human .beta.-galactoside-.alpha.-2,6-sialyltransferase I. Encompassed by the teachings as disclosed in here are homologous sialyltransferases comprising an amino acid motif corresponding to the motif from position 90 to position 108 in SEQ ID NO:1.

[0074] In a specific embodiment of all aspects as disclosed herein, the variant mammalian glycosyltransferase capable of catalyzing hydrolysis of the .alpha.2,6 glycosidic bond is a mammalian glycosyltransferase is derived, by way of amino acid deletion, from human .beta.-galactoside-.alpha.-2,6-sialyltransferase I according to SEQ ID NO:1, said sequence being truncated by a deletion from the N-terminus. In a further specific embodiment of all aspects as disclosed herein, the truncation deletion from the N-terminus is the contiguous sequence of position 1 to position 89 of SEQ ID NO:1.

[0075] Another aspect and a specific embodiment of all other aspects as disclosed herein is a fusion polypeptide comprising a polypeptide of a variant mammalian glycosyltransferase according to any embodiment as disclosed herein. A fusion protein or fusion polypeptide is a chimeric polypeptide comprising amino acid sequences of two or more polypeptides. The two or more polypeptides may have complementary functions, one of the polypeptides may provide a supplementary functional property, or one of the polypeptides may have a function unrelated to the others in the fusion polypeptide. One or more polypeptides comprising organelle targeting or retention sequences may be fused with a desired polypeptide to target the desired polypeptide to a specific cellular organelle, or retain the desired polypeptide within the cell. One or more polypeptides comprising a carrier sequence that aids in expression, purification and/or detection of the fusion polypeptide may be fused with a desired polypeptide (e.g., FLAG, a myc tag, a 6.times.His tag, GST fusions and the like). Particular fusion partners include N-terminal leader peptides capable of directing the variant mammalian glycosyltransferase portion of the fusion polypeptide to the secretory pathway of the host organism in which the fusion polypeptide is expressed. Thereby secretion in the extracellular space and the surrounding medium is facilitated. Yet, another aspect and a specific embodiment of all other aspects as disclosed herein is a nucleotide sequence encoding a variant mammalian glycosyltransferase according to any embodiment as disclosed herein or a fusion polypeptide comprising as a portion a variant mammalian glycosyltransferase according to any embodiment as disclosed herein. Importantly, the nucleotide sequence includes the sequence from position 90 to position 108 in SEQ ID NO:1, or the homologous equivalent thereof in the case of a sialyltransferase homologous to human .beta.-galacto side-.alpha.-2,6-sialyltransferase I.

[0076] Yet, another aspect and a specific embodiment of all other aspects as disclosed herein is an expression vector comprising a target gene and sequences facilitating expression of the target gene in a host organism transformed with the expression vector, wherein the target gene comprises a nucleotide sequence as disclosed herein.

[0077] Yet, another aspect and a specific embodiment of all other aspects as disclosed herein is a transformed host organism, wherein the host organism is transformed with an expression vector as disclosed herein. With particular advantage, Human Embryonic Kidney 293 (HEK) cells can be used to practice the teachings as disclosed in here. A particular advantage of these cells is that they are very suited targets for transfection followed by subsequent culture and transient expression of the target gene. Thus, HEK cells can be efficiently used to produce target proteins by way of recombinant expression. With great advantage, expression constructs are designed to direct the translation products to the secretory pathway leading to secretion of the variant mammalian glycosyltransferase or a fusion polypeptide as disclosed herein. Nevertheless, Jurkat, NIH3T3, HeLa, COS and Chinese Hamster Ovary (CHO) cells are well-known alternatives and are included herein as alternative host organisms for transformation and specific embodiments of all aspects as disclosed herein.

[0078] Yet, another aspect and a specific embodiment of all other aspects as disclosed herein is a method to produce recombinantly a variant mammalian glycosyltransferase, the method comprising the step of expressing in a host organism transformed with an expression vector a nucleotide sequence encoding a variant mammalian glycosyltransferase as disclosed herein, wherein a polypeptide is formed, thereby producing variant mammalian glycosyltransferase.

[0079] According to earlier knowledge, N-terminally truncated variants of glycosyltransferases are advantageously used in vitro due to their lack of transmembrane domains. Thus, such variants are useful for catalyzing and performing glycosyltransferase reactions in solution. It was surprisingly found and is disclosed herein that particularly the N-terminally truncated variant .DELTA.89 hST6Gal-I displays different activities in vitro, e.g. when incubated with glycosylated antibodies and in the presence of 5'-CMP. Thus, a specific embodiment of the present disclosure and all aspects and embodiments herein is a variant mammalian glycosyltransferase capable of catalyzing hydrolysis of a .alpha.2,6 glycosidic bond of a N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine moiety of a bi-sialylated glycoprotein, i.e. a glycoprotein comprising two separate terminal N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine moieties in one or more glycan portion(s) of the glycoprotein. In a specific embodiment, only one .alpha.2,6 glycosidic bond in a is hydrolyzed. In a further specific embodiment, the bi-sialylated glycoprotein is a bi-sialylated IgG immunoglobulin.

[0080] As an exemplary case, the IgG-Fc glycan G2 has two galactose moieties at the termini of the antennate branches which can be sialylated. Under suitable reaction conditions, the N-terminally truncated variant .DELTA.89 hST6Gal-I catalyzes the synthesis of IgG with bi-sialylated G2 glycans (G2+2SA) at the immunoglobulin Fc portion. However, upon accumulation of 5'-CMP the enzyme variant acts as a sialidase catalyzing removal by hydrolysis of a sialic acid moiety from the bi-sialylated (G2+2SA) antibodies resulting in mono-sialylated (G2+1SA) antibodies. This property was found unexpectedly and appears to represent an intrinsic sialidase (neuraminidase) activity.

[0081] In a basic publication on human ST6Gal-I it was stated that the enzyme does not contain any sialidase activity, see Sticher et al. Glycoconjugate Journal 8 (1991) 45-54. In view of the present surprising finding it becomes possible to preferentially synthesize glycoproteins with mono-, bi- or even higher sialylated glycans, using the same enzyme and controlling the reaction kinetics of the enzyme by controlling CMP in the sialylation reaction mixture. A further advantage is that both activities, sialylation activity and sialidase activity are provided by the same enzyme, in the same reaction vessel.

[0082] The general finding documented in the present disclosure is, however, that there exists a variant mammalian glycosyltransferase, specifically a glycosyltransferase according to the present disclosure, which is capable of catalyzing hydrolysis of the .alpha.2,6 glycosidic bond of a N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-- .beta.-D-glucosamine moiety of a glycan in a glycoprotein. In addition to the already known sialyltransferase (sialylation) activity the surprising finding was that at least as specifically shown for the N-terminally truncated human .beta.-galactoside-.alpha.-2,6-sialyltransferase I having the amino acid sequence of SEQ ID NO:2 there is not only conventional sialyltransferase but also sialidase enzymatic activity mediated by this enzyme. Interestingly, in the exemplary cases these two activities were not observed at the same time, which may partly explain the unexpected finding. Thus, in the absence of CMP sialyltransferase activity dominates in the beginning and sialidase activity becomes apparent only at a later stage during the incubation, once sufficient amounts of CMP have accumulated. Nevertheless, the apparent recognition of two distinct activities of the same enzyme allows to control the extent of sialylation of target molecules, e.g. by way of varying incubation time.

[0083] However, in a very elegant way, sialylation can be maximized by adding to the sialylation reaction mixture an enzyme capable of hydrolyzing the phosphoester bond in 5'-cytidine-monophosphate under the conditions which permit sialyltransferase enzymatic activity. Thus, the by-product CMP is removed and the sialylation catalysis by the sialyltransferase is not counteracted.

[0084] Yet, another aspect and a specific embodiment of all other aspects as disclosed herein is the use of an enzyme capable of hydrolyzing the phosphoester bond in 5'-cytidine-monophosphate under the conditions which permit sialyltransferase enzymatic activity to maintain glycosyltransferase activity and/or inhibit sialidase activity of a variant mammalian glycosyltransferase as disclosed herein, specifically the N-terminally truncated human .beta.-galactoside-.alpha.-2,6-sialyltransferase I having the amino acid sequence of SEQ ID NO:2.

[0085] Such controlled sialylation is provided as a novel means to synthesize in vitro mono-, bi-, and higher sialylated glycoproteins with a desired degree of sialylation. Thus, though exemplified by showing the desired technical effects with IgG molecules, the uses according to the disclosures in here also allow to process other glycoproteins in a similar way, with the proviso that concerning glysosyltransferase activity, the glycoproteins comprise two or more terminal antennate .beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-glucosamine moieties. The same reasoning applies in an analogous way to glycolipids.

[0086] In a particular example, recombinant humanized IgG1 and IgG4 monoclonal antibodies (mabs), characterized as G2+0SA (=two acceptor sites present, no sialylation at any acceptor site), as well as EPO (=erythropoietin) were used as targets in sialylation experiments (30 .mu.g enzyme/300 .mu.g target protein). .DELTA.89 hST6Gal-I was used under standard reaction conditions and the G2+0SA, G2+1SA (=mono-sialylation, one out of two acceptor sites is sialylated) and G2+2SA (=bi-sialylation, both possible acceptor sites are sialylated) status was analyzed by mass spectrometry.

[0087] Due to the high expression rates and the efficient purification procedures the exemplary .DELTA.89 hST6Gal-I but also functionally equivalent enzymes can be made available in large quantities and with high purity. The variant .DELTA.89 hST6Gal-I enzyme is active with high molecular weight substrates of which monoclonal antibodies are just one example. Depending on the incubation time, .DELTA.89 hST6Gal-I in combination with a CMP-hydrolyzing enzyme shows good performance in sialylation experiments using monoclonal antibodies with bi-antennary glycan as substrate. Using embodiments of the present disclosure the preferably bi-sialylated glycans are obtained with great advantage after shorter incubation periods, such as 8 hours. Tetra-antennary glycans are also accepted as substrate (data not shown). The results demonstrate technical advantage for in vitro glycoengineering of therapeutic antibodies.

[0088] The following items further provide specific aspects of the disclosure, and specific embodiments to practice the teachings provided herein. [0089] 1. Use of a soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprising the amino acid motif from position 90 to position 108 in SEQ ID NO:1 for in vitro hydrolyzing in the presence of 5'-cytidine-monophosphate the .alpha.2,6 glycosidic bond in a N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-g- lucosamine moiety, the moiety being a terminal structure of a glycan in a sialylated glycoprotein or glycolipid. [0090] 2. The use according to item 1, wherein the soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprises the amino acids from position 90 to position 406 in SEQ ID NO:1. [0091] 3. The use according to any of the items 1 and 2, wherein the amino acid sequence of the soluble .beta.-galactoside-.alpha.-2,6-sialyltransferase I is the amino acid sequence of SEQ ID NO:2. [0092] 4. The use according to any of the items 1 to 3, wherein the glycoprotein is selected from the group consisting of a cell surface glycoprotein and a serum glycoprotein. [0093] 5. The use according to item 4, wherein the serum glycoprotein is selected from a glycosylated protein signaling molecule, a glycosylated immunoglobulin, and a glycosylated protein of viral origin. [0094] 6. The use according to any of the items 1 to 5, wherein the glycoprotein is recombinantly produced. [0095] 7. The use according to item 6, wherein the glycoprotein is recombinantly produced in a transformed host cell of mammalian origin. [0096] 8. The use according to any of the items 1 to 7, wherein the glycoprotein is an immunoglobulin of human origin or a humanized immunoglobulin, the immunoglobulin being selected from the group consisting of IgG1, IgG2, IgG3, IgG4. [0097] 9. The use according to any of the items 1 to 7, wherein the glycoprotein is selected from EPO and asialofetuin. [0098] 10. An aqueous composition comprising [0099] (a) a soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprising the amino acid motif from position 90 to position 108 in SEQ ID NO:1; [0100] (b) cytidine-5'-monophospho-N-acetylneuraminic acid, or a functional equivalent thereof; [0101] (c) a glycosylated target molecule selected from a glycoprotein and a glycolipid, the target molecule comprising one or more antenna(e), at least one antenna having as a terminal structure a .beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-glucosamine moiety with a hydroxyl group at the C6 position in the galactosyl residue; [0102] (d) an aqueous solution permitting glycosyltransferase enzymatic activity; [0103] wherein the aqueous composition further comprises an enzyme capable of hydrolyzing the phosphoester bond in 5'-cytidine-monophosphate under conditions permitting glycosyltransferase enzymatic activity. [0104] 11. The aqueous composition according to item 10, wherein the soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprises the amino acids from position 90 to position 406 in SEQ ID NO:1. [0105] 12. The aqueous composition according to any of the items 10 and 11, wherein the amino acid sequence of the soluble .beta.-galactoside-.alpha.-2,6-sialyltransferase I is the amino acid sequence of SEQ ID NO:2. [0106] 13. The aqueous composition according to any of the items 10 to 12, wherein the glycosylated target molecule is a glycoprotein selected from the group consisting of a cell surface glycoprotein and a serum glycoprotein. [0107] 14. The aqueous composition according to any of the items 10 to 13, wherein the serum glycoprotein is selected from a glycosylated protein signaling molecule, a glycosylated immunoglobulin, and a glycosylated protein of viral origin. [0108] 15. The aqueous composition according to any of the items 10 to 14, wherein the glycoprotein is recombinantly produced. [0109] 16. The aqueous composition according to item 15, wherein the glycoprotein is recombinantly produced in a transformed host cell of mammalian origin. [0110] 17. The aqueous composition according to any of the items 10 to 16, wherein the glycoprotein is an immunoglobulin of human origin or a humanized immunoglobulin, the immunoglobulin being selected from the group consisting of IgG1, IgG2, IgG3, IgG4. [0111] 18. The aqueous composition according to any of the items 10 to 16, wherein the glycoprotein is selected from EPO and asialofetuin. [0112] 19. The aqueous composition according to any of the items 10 to 18, wherein the aqueous solution comprises water, a buffer salt capable of buffering in the pH range of pH 6 to pH 8, and optionally a compound selected from the group consisting of a neutral salt, a salt with a divalent cation, an antioxidant, a surfactant and a mixture thereof. [0113] 20. The aqueous composition according to any of the items 10 to 19, the composition having a temperature of 0.degree. C. to 40.degree. C. [0114] 21. The aqueous composition according to any of the items 10 to 20, wherein the enzyme capable of the hydrolyzing phosphoester bond in 5'-cytidine-monophosphate is selected from the group consisting of an alkaline phosphatase, an acid phosphatase, and a 5' nucleotidase. [0115] 22. The aqueous composition according to item 21, wherein the alkaline phosphatase is selected from the group consisting of alkaline phosphatase of bacterial origin, shrimp alkaline phosphatase, calf intestine alkaline phosphatase, human placental alkaline phosphatase, and a mixture thereof. [0116] 23. The aqueous composition according to item 22, wherein the aqueous composition further comprises Zn.sup.2+ ions. [0117] 24. The aqueous composition according to item 21, wherein the 5' nucleotidase is 5'nucleotidase CD73 of mammalian origin, specifically of human origin. [0118] 25. Use of an aqueous composition according to any of the items 10 to 24 for reducing 5'-cytidine-monophosphate-mediated inhibition and thereby maintaining the sialylating activity of the soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprising the amino acid motif from position 90 to position 108 in SEQ ID NO:1. [0119] 26. The use according to item 25, wherein the sialylating activity catalyzes transfer and covalent coupling of the sialic acid residue, or the functional equivalent thereof, from the co-substrate to a .beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-glucosamine moiety with a hydroxyl group at the C6 position in the galactosyl residue, the moiety being a terminal structure of a glycan of a glycosylated target molecule selected from a glycoprotein and a glycolipid. [0120] 27. A method of producing in vitro a sialylated target molecule, the method comprising the steps of [0121] (a) providing an aqueous composition according to any of the items 10 to 24; [0122] (b) forming one or more terminal antennal N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.- beta.-D-glucosamine residue(s) by incubating the aqueous composition of step (a), thereby reacting cytidine-5'-monophospho-N-acetylneuraminic acid, or a functional equivalent thereof, as co-substrate, thereby forming 5'-cytidine-monophosphate; [0123] (c) hydrolyzing the phosphoester bond of the 5'-cytidine-monophosphate formed in step (b), thereby reducing 5'-cytidine-monophosphate-mediated inhibition, thereby maintaining the activity of the soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I; [0124] thereby producing in vitro the sialylated target molecule. [0125] 28. The method according to item 27, wherein the method is performed at a temperature of 0.degree. C. to 40.degree. C. [0126] 29. The method according to any of the items 27 and 28, wherein steps (b) and (c) are performed in the same vessel. [0127] 30. The method according to any of the items 27 to 29, wherein steps (b) and (c) are performed for a period selected from the group consisting of 2 h to 96 h, 2 h to 23 h, 2 h to 6 h, and about 2 h. [0128] 31. The method according to any of the items 27 to 29, wherein steps (b) and (c) are performed for a period selected from the group consisting of 6 h to 96 h, 6 h to 23 h, and about 6 h. [0129] 32. The method according to any of the items 27 to 29, wherein steps (b) and (c) are performed for a period selected from the group consisting of 23 h to 96 h, and about 23 h. [0130] 33. The method according to any of the items 27 to 29, wherein steps (b) and (c) are performed for a period of about 96 h. [0131] 34. Use of a soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I lacking the amino acid motif from position 90 to position 108 in SEQ ID NO:1 for transferring in vitro and in the presence of 5'-cytidine-monophosphate a 5-N-acetylneuraminic acid residue from the donor compound cytidine-5'-monophospho-N-acetylneuraminic acid, or from a functional equivalent thereof, to an acceptor, the acceptor being terminal .beta.-D-galactosyl-1,4-N-acetyl-3-D-glucosamine in a glycan moiety of a glycoprotein or a glycolipid. [0132] 35. The use according to item 34, wherein the soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprises the amino acids from position 109 to position 406 in SEQ ID NO:1. [0133] 36. The use according to any of the items 34 and 35, wherein the amino acid sequence of the soluble .beta.-galactoside-.alpha.-2,6-sialyltransferase I is the amino acid sequence of SEQ ID NO:5. [0134] 37. The use according to any of the items 34 to 36, wherein the glycoprotein is selected from the group consisting of a cell surface glycoprotein and a serum glycoprotein. [0135] 38. The use according to item 37, wherein the serum glycoprotein is selected from a glycosylated protein signaling molecule, a glycosylated immunoglobulin, and a glycosylated protein of viral origin. [0136] 39. The use according to any of the items 34 to 38, wherein the glycoprotein is recombinantly produced. [0137] 40. The use according to item 39, wherein the glycoprotein is recombinantly produced in a transformed host cell of mammalian origin. [0138] 41. The use according to any of the items 34 to 40, wherein the glycoprotein is an immunoglobulin of human origin or a humanized immunoglobulin, the immunoglobulin being selected from the group consisting of IgG1, IgG2, IgG3, IgG4. [0139] 42. The use according to any of the items 34 to 40, wherein the glycoprotein is selected from EPO and asialofetuin. [0140] 43. An aqueous composition comprising [0141] (a) a soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I lacking the amino acid motif from position 90 to position 108 in SEQ ID NO:1; [0142] (b) cytidine-5'-monophospho-N-acetylneuraminic acid, or a functional equivalent thereof; [0143] (c) a glycosylated target molecule selected from a glycoprotein and a glycolipid, the target molecule comprising one or more antenna(e), at least one antenna having as a terminal structure a .beta.-D-galactosyl-1,4-N-acetyl-.beta.-D-glucosamine moiety with a hydroxyl group at the C6 position in the galactosyl residue; [0144] (d) an aqueous solution permitting glycosyltransferase enzymatic activity; [0145] wherein the aqueous composition further comprises 5'-cytidine-monophosphate. [0146] 44. The aqueous composition according to item 43, wherein the soluble human .beta.-galactoside-.alpha.-2,6-sialyltransferase I comprises the amino acids from position 109 to position 406 in SEQ ID NO:1. [0147] 45. The aqueous composition according to any of the items 43 and 44, wherein the amino acid sequence of the soluble .beta.-galactoside-.alpha.-2,6-sialyltransferase I is the amino acid sequence of SEQ ID NO:5. [0148] 46. The aqueous composition according to any of the items 43 to 45, wherein the glycosylated target molecule is a glycoprotein selected from the group consisting of a cell surface glycoprotein and a serum glycoprotein. [0149] 47. The aqueous composition according to any of the items 43 to 46, wherein the serum glycoprotein is selected from a glycosylated protein signaling molecule, a glycosylated immunoglobulin, and a glycosylated protein of viral origin. [0150] 48. The aqueous composition according to any of the items 43 to 47, wherein the glycoprotein is recombinantly produced. [0151] 49. The aqueous composition according to item 48, wherein the glycoprotein is recombinantly produced in a transformed host cell of mammalian origin. [0152] 50. The aqueous composition according to any of the items 43 to 49, wherein the glycoprotein is an immunoglobulin of human origin or a humanized immunoglobulin, the immunoglobulin being selected from the group consisting of IgG1, IgG2, IgG3, IgG4. [0153] 51. The aqueous composition according to any of the items 43 to 49, wherein the glycoprotein is selected from EPO and asialofetuin. [0154] 52. The aqueous composition according to any of the items 43 to 51, wherein the aqueous solution comprises water, a buffer salt capable of buffering in the pH range of pH 6 to pH 8, and optionally a compound selected from the group consisting of a neutral salt, a salt with a divalent cation, an antioxidant, a surfactant and a mixture thereof. [0155] 53. The aqueous composition according to any of the items 43 to 52, the composition having a temperature of 0.degree. C. to 40.degree. C. [0156] 54. A method of producing in vitro a sialylated target molecule, the method comprising the steps of [0157] (a) providing an aqueous composition according to any of the items 43 to 53; [0158] (b) forming one or more terminal antennal N-acetylneuraminyl-.alpha.2,6-.beta.-D-galactosyl-1,4-N-acetyl-.- beta.-D-glucosamine residue(s) by incubating the aqueous composition of step (a), thereby reacting cytidine-5'-monophospho-N-acetylneuraminic acid, or a functional equivalent thereof, as co-substrate, thereby forming 5'-cytidine-monophosphate; [0159] (c) accumulating 5'-cytidine-monophosphate formed in step (b); [0160] thereby producing in vitro the sialylated target molecule. [0161] 55. The method according to item 54, wherein the method is performed at a temperature of 0.degree. C. to 40.degree. C. [0162] 56. The method according to any of the items 54 and 55, wherein steps (b) and (c) are performed in the same vessel. [0163] 57. The method according to any of the items 54 to 56, wherein steps (b) and (c) are performed for a period selected from the group consisting of 2 h to 72 h. [0164] 58. Use of an enzyme capable of the hydrolyzing phosphoester bond in 5'-cytidine-monophosphate for maintaining sialyltransferase enzymatic activity in a composition according to any of the items 10 to 24. [0165] 59. Use of an enzyme capable of the hydrolyzing phosphoester bond in 5'-cytidine-monophosphate for inhibiting sialidase enzymatic activity in a composition according to any of the items 10 to 24. [0166] 60. A preparation of sialylated immunoglobulins, each immunoglobulin having a plurality of acceptor sites for human .beta.-galactoside-.alpha.-2,6-sialyltransferase I, wherein less than 25% of the acceptor sites in the preparation of sialylated immunoglobulins are not sialylated, and 75% or more are sialylated, wherein the preparation is obtained by a method according to any of the items 27 to 33.

[0167] 61. The preparation according to item 60, wherein less than 20% of the acceptor sites in the preparation of sialylated immunoglobulins are not sialylated, and 80% or more are sialylated. [0168] 62. The preparation according to item 60, wherein less than 10% of the acceptor sites in the preparation of sialylated immunoglobulins are not sialylated, and 90% or more are sialylated.

[0169] The Examples that follow are illustrative of specific embodiments of the disclosure, and various uses thereof. They set forth for explanatory purposes only, and are not to be taken as limiting the disclosure.

Example 1

Test for Sialyltransferase Enzymatic Activity

[0170] Asialofetuin (desialylated fetuin, Roche Applied Science) was used as acceptor and CMP-9-fluoro-NANA (CMP-9-fluoresceinyl-NeuAc) was used as donor substrate (Brossmer, R. & Gross H. J. (1994) Meth. Enzymol. 247, 177-193). Enzymatic activity of a sialyltransferase was determined by measuring the transfer of sialic acid from the donor compound to asialofetuin. The reaction mix (35 mM MES, pH 6.0, 0.035% Triton X-100, 0.07% BSA) contained 2.5 .mu.g of enzyme sample, 5 .mu.L asialofetuin (20 mg/mL) and 2 .mu.L CMP-9-fluoro-NANA (1.0 mg/mL) in a total volume of 51 .mu.L. The reaction mix was incubated at 37.degree. C. for 30 min. The reaction was stopped by the addition of 10 .mu.L of the inhibitor CTP (10 mM). The reaction mix was loaded onto a PD10 desalting column equilibrated with 0.1 M Tris/HCl, pH 8.5. Fetuin was eluted from the column using the equilibration buffer. The fractions size was 1 mL. The concentration of formed fetuin was determined using a fluorescence spectrophotometer. Excitation wave length was 490 nm, emission was measured at 520 nm. Enzymatic activity was expressed as RFU (relative fluorescence unit). 10,000 RFU/.mu.g is equivalent to a specific activity of 0.0839 nmol/.mu.g.times.min.

Example 2

SDS Gel Electrophoresis

[0171] Analytical SDS gel electrophoresis was carried out using NuPAGE gels (4-12%, Invitrogen). Samples (36 .mu.L) were diluted with 12 .mu.L NuPAGE LDS sample buffer (Invitrogen) and incubated for 2 min at 85.degree. C. Aliquots, typically containing 5 .mu.g protein were loaded on the gel. The gels were stained using SimplyBlue SafeStain (Invitrogen).

Example 3

N-Terminal Sequencing by Edman Degradation

[0172] The N-terminal sequences of expressed variants of human ST6Gal-I were analyzed by Edman degradation using reagents and devices obtained from Life Technologies. Preparation of the samples was done as described in the instruction manual of the Life Technologies ProSorb Sample Preparation cartridges (catalogue number 401950) and the Life Technologies ProBlott Mini PK/10 membranes (catalogue number 01194). For sequencing the Procise Protein Sequencing Platform was used.

Example 4

Mass Spectrometry of Glycosylated Human ST6Gal-I Enzymes

[0173] The molecular masses of variants of human ST6Gal-I expressed in HEK cells were analyzed. Glycosylated forms of human ST6Gal-I were prepared, and prepared material was analyzed using Micromass Q-Tof Ultima and Synapt G2 HDMS devices (Waters UK) and MassLynx V 4.1 software.

[0174] For mass spectrometry measurement the samples were buffered in electrospray medium (20% acetonitrile+1% formic acid). The buffer exchange was performed with Illustra.TM. MicroSpin.TM. G-25 columns (GE-Healthcare). 20 .mu.g sialyltransferase variant with a concentration of 1 mg/mL was applied to the pre-equilibrated column and eluated by centrifugation. The resulting eluate was analyzed by electrospray ionization mass spectrometry.

Example 5

Mass Spectrometry of Deglycosylated Human ST6Gal-I Enzymes

[0175] The molecular masses of variants of human ST6Gal-I expressed in HEK cells were analyzed. Delycosylated forms of human ST6Gal-I were analyzed using Micromass Q-Tof Ultima and Synapt G2 HDMS devices (Waters UK) and MassLynx V 4.1 software.

[0176] For deglycosylation samples of the sialyltransferase were denatured and reduced. To 100 .mu.g sialyltransferase 45 .mu.L denaturing buffer (6 M guanidinium hydrochloride) and 13 .mu.L TCEP (=tris(2-carboxyethyl)phosphine; 0.1 mM, diluted in denaturing buffer) were added. Further the appropriate volume of ultrapure water was added, so that the overall concentration of guanidinium hydrochloride was about 4 M. After incubation of the sample for 1 h at 37.degree. C. the buffer was changed using a Bio-SpinR 6 Tris column (Bio Rad), which was pre-equilibrated with ultrapure water. The whole sample was applied onto the column and eluted by centrifugation. To the resulting eluate 5.5 .mu.L of 0.1 U/.mu.L solution of PNGase-F was added and incubated at 37.degree. C. over night. Afterwards the samples were adjusted to 30% ACN (=acetonitrile) and 1% FA (=formamide) and analyzed by electrospray ionization mass spectrometry.

Example 6

[0177] Cloning of pM1MT Expression Constructs for Transient Expression in Mammalian Host Cells of Truncated Variant .DELTA.89 of Human ST6Gal-I

[0178] Truncated variant .DELTA.89 of human ST6Gal-I was cloned for transient expression using an Erythropoietin signal peptide sequence (Epo) and a peptide spacer of two amino acids ("AP").

[0179] For the Epo-AP-.DELTA.89 hST6Gal-I construct codon-optimized cDNAs were synthesized, see SEQ ID NO:3. Instead of the natural leader sequences and the N-terminal protein sequences, the hST6Gal-I coding region harbors the Erythropoietin signal sequence plus AP linker sequence in order to ensure correct processing of expressed polypeptides by the secretion machinery of the host cell line. In addition, the expression cassettes features SalI and BamHI restriction sites for cloning into the multiple cloning site of the predigested pM1MT vector fragment (Roche Applied Science). Expression of the ST6Gal-I coding sequence is therefore under control of a human cytomegalovirus (CMV) immediate-early enhancer/promoter region, followed by an "intron A" for regulated expression, and a BGH polyadenylation signal.

[0180] Expression of the Epo-AP-.DELTA.89 hST6Gal-I construct in HEK cells, and secretion of .DELTA.89 hST6Gal-I protein into cell supernatant was performed as described in Example 8.

Example 7

[0181] Cloning of pM1MT Expression Constructs for Transient Expression in Mammalian Host Cells of Truncated Variant .DELTA.108 of Human ST6Gal-I

[0182] Truncated variant .DELTA.108 of human ST6Gal-I was cloned for transient expression using an Erythropoietin signal peptide sequence (Epo) and a peptide spacer of four amino acids ("APPR"). For the Epo-APPR-.DELTA.108 hST6Gal-I construct a codon-optimized cDNAs was synthesized, see SEQ ID NO:6. The natural hST6Gal-I-derived mRNA leader and N-terminal protein sequences were exchanged with the Erythropoetin signal sequence and the "APPR" linker sequence to ensure correct processing of the polypeptide by the secretion machinery of the HEK host cell line. In addition, the expression cassettes feature SalI and BamHI sites for cloning into the multiple cloning site of the pre-digested pM1MT vector fragment (Roche Applied Science). Expression of the hST6Gal-I coding sequence was thereby put under the control of a human cytomegalovirus (CMV) immediate-early enhancer/promoter region; the expression vector further featured an "intron A" for regulated expression and a BGH polyadenylation signal. Expression of the Epo-APPR-.DELTA.108 hST6Gal-I construct (SEQ ID NO:6) in HEK cells, and secretion of .DELTA.108 hST6Gal-I protein into cell supernatant was performed as described in Example 8.

Example 8

Transformation HEK Cells and Transient Expression and Secretion

[0183] Transient gene expression (TGE) by transfection of plasmid DNA is a rapid strategy to produce proteins in mammalian cell culture. For high-level expression of recombinant human proteins a TGE platform based on a suspension-adapted human embryonic kidney (HEK) 293 cell line was used. Cells were cultured in shaker flasks at 37.degree. C. under serum-free medium conditions. The cells were transfected at approx. 2.times.10.sup.6 vc/mL with the pM1MT expression plasmids (0.5 to 1 mg/L cell culture) complexed by the 293-Free.TM. (Merck) transfection reagent according to the manufacturer's guidelines. Three hours post-transfection, valproic acid, a HDAC inhibitor, was added (final conc. 4 mM) in order to boost the expression (Backliwal et al. (2008), Nucleic Acids Research 36, e96). Each day, the culture was supplemented with 6% (v/v) of a soybean peptone hydrolysate-based feed. The culture supernatant was collected at day 7 post-transfection by centrifugation.

Example 9

[0184] Purification of the N-Terminal Truncation Variants of Human ST6Gal-I from Supernatants of Transformed HEK Cells

[0185] HEK cells were transformed as described in Example 8. Expression constructs were prepared as described in Examples 6 and 7.

[0186] From supernatants of HEK cell fermentations the two enzyme variants Epo-AP-.DELTA.89 hST6Gal-I and Epo-APPR-.DELTA.108 hST6Gal-I were purified using a simplified purification protocol. In a first step, a volume of 0.1 L of culture supernatant was filtrated (0.2 .mu.m), and the solution was dialysed against buffer A (20 mM potassium phosphate, pH 6.5). The dialysate was loaded onto a S-Sepharose.TM. ff (Fast Flow) column (1.6 cm.times.2 cm) equilibrated with buffer A. After washing with 100 mL buffer A, the enzyme was eluted with a linear gradient of 10 mL buffer A and 10 mL of buffer A with 200 mM NaCl, followed by a wash step using 48 mL of buffer A with 200 mM NaCl. Fractions (4 mL) were analysed by an analytical SDS gel electrophoresis.

[0187] Fractions containing the .DELTA.89 hST6Gal-I enzyme were pooled and dialyzed against buffer B (50 mM MES, pH 6.0). The dialyzed pool was loaded onto a Heparin Sepharose ff column (0.5 cm.times.5 cm) equilibrated with buffer B and eluted using buffer B with 200 mM NaCl. Fractions (1 mL) containing the enzyme were pooled and dialyzed against buffer B. Protein concentrations were determined at 280 nm (E280 nm [1 mg/mL]=1.931). Mass spectrometry analysis showed that the recombinantly expressed Epo-AP-.DELTA.89 hST6Gal-I enzyme was secreted without the N-terminal amino acids AP. This finding was unexpected and indicated unusual cleavage of the expressed protein by the signal peptidase while removing the Epo portion. For the recombinant human .DELTA.89 hST6Gal-I enzyme a specific activity of 3.75 nmol/.mu.g.times.min was determined. FIG. 2 shows the results of a SDS-PAGE of recombinant .DELTA.89 hST6Gal-I variant purified from HEK cells.

[0188] Fractions containing the .DELTA.108 hST6Gal-I enzyme were pooled and dialyzed against storage buffer (20 mM potassium phosphate, 100 mM sodium chloride, pH 6.5). Protein concentration was determined at a wave length of 280 nm using a molar extinction coefficient of 1.871. Mass spectrometric analysis of the recombinant protein secreted from the HEK cells transformed with the Epo-APPR-.DELTA.108-hST6Gal-I expression construct confirmed the N-terminal sequence "APPR", thus indicating the expected cleavage of the EPO signal sequence by the signal peptidase. For the recombinant human .DELTA.108 hST6Gal-I variant from HEK cells a specific activity of >600 RFU/.mu.g was determined. FIG. 3 shows the results of a SDS-PAGE of recombinant .DELTA.108 hST6Gal-I variant purified from HEK cells.

Example 10

[0189] Sialylation of Humanized Monoclonal Antibody IgG4 MAB Using .DELTA.89 hST6Gal-I

[0190] A highly galactosylated humanized monoclonal antibody IgG4 MAB was used in sialylation experiments. The reaction mixture contained IgG4 MAB (300 .mu.g in 55 .mu.L 35 mM sodium acetate/Tris buffer pH 7.0), the donor substrate CMP-NANA (150 .mu.g in 50 .mu.L water) and sialyltransferase (30 .mu.g .DELTA.89 hST6Gal-I in 20 mM potassium phosphate, 0.1 M NaCl, pH 6.5). The samples were incubated at 37.degree. C. for a defined time. To stop the reaction the samples were frozen at -20.degree. C. For mass analysis 100 .mu.L denaturing buffer (6 M guanidinium chloride) and 30 .mu.L TCEP (=tris(2-carboxyethyl)phosphine; 0.1 mM, diluted in denaturing buffer) were added to the samples and the samples were incubated at 37.degree. C. for 1 h. The samples were buffered in electrospray medium [20% ACN (=acetonitrile), 1% FA (=formamide)] using pre-equilibrated Illustra.TM. Nap5-Columns (GE-Healthcare). Samples were analyzed by electrospray ionization mass spectrometry and the content of G2+0SA, G2+1SA and G2+2SA N-glycans was determined. A Micromass Q-Tof Ultima and a Synapt G2 HDMS device (Waters UK) were used, the software used was MassLynx V 4.1. To determine the kinetics of the sialylation the reaction was incubated up to 72 h. FIG. 4 shows the relative amounts of differently sialylated target proteins obtained after different time points during the incubation period.

[0191] The content of G2+0SA, G2+1SA and G2+2SA was determined by mass spectrometry. For the variant .DELTA.89 hST6Gal-I already after 2 h of incubation a high content (88%) of the bi-sialylated form G2+2SA was obtained, see FIG. 4. The data also show that the content of G2+0SA and G2+1SA again increased over time due to the intrinsic CMP-dependent sialidase (neuraminidase) activity of .DELTA.89 hST6Gal-I. After an incubation of 48 h a G2+1SA content of 71% was obtained.

[0192] FIG. 5 shows the spectra obtained by mass spectrometric analysis of different samples of IgG4 MAB. Samples were taken at time point t=0 (lower panel), time point t=8 h (middle panel) and time point t=48 h (upper panel). The mass over charge (m/z) signals of one charge state in the mass spectrum of the IgG molecule with G2+0SA, G2+1SA and G2+2SA glycans are depicted. The relative intensities of the different sialylated species are derived from these signals. Corresponding to FIG. 4, at t=0 h G2+0SA is the major glycan species. At t=8 h the signal for G2+2SA is the dominant form whereas at t=48 h, G2+1SA is the most abundant species. For the determined numerical values see FIG. 4.

Example 11

[0193] Inhibition of Sialidase Activity of .DELTA.89 hST6Gal-I by CTP

[0194] The compound cytidine-5'-triphosphate (CTP) is a known potent inhibitor of sialyltransferases (Scudder P R & Chantler E N BBA 660 (1981) 136-141). To demonstrate that the sialidase activity is an intrinsic activity of .DELTA.89 hST6Gal-I, inhibition experiments were performed. In the first phase of the experiment the sialylation of IgG4 MAB by .DELTA.89 hST6Gal-I was performed to achieve a high content of G2+2SA (see Example 10). After 7 h of incubation the G2+2SA content was 94%. Subsequently, CTP was added to inhibit the sialidase activity of .DELTA.89 hST6Gal-I (final concentration of CTP: 0.67 mM). At different times samples were taken and the content of G2+0SA, G2+1SA and G2+2SA was determined by mass spectrometry. The results are shown in FIG. 6. Compared to inhibitor-free conditions shown in FIG. 4 the degradation of G2+2SA caused by the sialidase activity was significantly reduced. After 72 h of incubation 73% of G2+2SA were still present. The inhibition of the sialidase activity by a known inhibitor of sialyltransferase activity strongly indicates that both activities are located in the same active center of .DELTA.89 hST6Gal-I.

Example 12

[0195] Sialylation of IgG1 MAB with .DELTA.89 hST6Gal-I

[0196] The amount of 15 mg of a highly galactosylated humanized monoclonal antibody IgG1 MAB was used for sialylation treatment. The reaction mixture contained defined amounts of IgG1 MAB (15 mg in 1,854 .mu.L aqueous buffer containing 20 mM sodium acetate, 50 mM Tris buffer, pH 8.0), the donor substrate CMP-NANA (7,500 .mu.g in 2,500 .mu.L water) and sialyltransferase (1,500 .mu.g recombinantly produced and purified .DELTA.89 hST6Gal-I in 202 .mu.L of 20 mM potassium phosphate, 0.1 M NaCl, pH 6.5). The components were mixed and the resulting reaction mixture was incubated at 37.degree. C. for different times, including 2 h, 4 h, 8 h, 24 h, and 48 h. Purification of sialylated antibody was performed as in Example 13.

[0197] To analyze the degree of sialylation, 124 .mu.L denaturing buffer (6 M Guanidinium chloride in water) and 30 .mu.L TCEP (=tris(2-carboxyethyl)phosphine; 0.1 mM, diluted in denaturing buffer) were added to 76 .mu.L (corresponding to 250 .mu.g IgG1 MAB) and the sample was incubated at 37.degree. C. for 1 h. After that the sample was buffered in electrospray medium [20% ACN (=acetonitrile), 1% FA (=formamide)] using pre-equilibrated Illustra.TM. Nap5-Columns (GE-Healthcare). Subsequently the sample was analyzed by electrospray ionization mass spectrometry, and the content of G2+0SA, G2+1SA and G2+2SA N-glycans was determined. A Synapt G2 HDMS device (Waters UK) was used, the software used was MassLynx V 4.1.

Example 13

Purification of Sialylated IgG1 MAB

[0198] To remove sialyltransferase and CMP-NANA from the incubated sialylation reaction mixture of Example 12, incubated IgG1 MAB was purified using Protein A. The reaction mixture was applied to a Protein A column equilibrated with 25 mM Tris, 25 mM NaCl, 5 mM EDTA (=ethylenediaminetetraacetic acid), pH 7. The column was washed with 25 mM Tris, 25 mM NaCl, 500 mM TMAC (=tetramethylammonium chloride), 5 mM EDTA pH 5.0 and then with 25 mM Tris, 25 mM NaCl, 5 mM EDTA pH 7.1. IgG1 MAB was eluted with 25 mM Na-Citrate. To avoid spontaneous desilylation at low pH, the pH was adjusted to pH 7.0 using 1 M Tris pH 9.0. Using this procedure sialylated IgG1 MAB was obtained in pure form, with a typical yield of 12 mg.

Example 14

[0199] Sialidase Activity of .DELTA.89 hST6Gal-I on IgG1 MAB in the Presence and Absence of CMP

[0200] Cytidine monophosphate (5'-CMP, =CMP) is a product of the reaction catalyzed by sialyltransferase enzymes, generated in the course of the glycosyltransferase reaction from the co-substrate CMP-NANA. With incubation time of a sialylation reaction CMP accumulates in the reaction mixture. To demonstrate that the inherent sialidase activity is CMP-dependent, highly sialylated IgG1 monoclonal antibody IgG1 MAB G2+2SA was prepared by incubation with .DELTA.89 hST6Gal-I in the presence of CMP-NANA, as described in Example 12, and purified as described in Example 13.

[0201] To an amount of 1,250 .mu.g (in 194 .mu.L) highly sialylated IgG1 MAB according to Example 12 with an incubation period for sialylation of 8 h, 125 .mu.g sialyltransferase variant (30 .mu.g/300 .mu.g IgG1 MAB) was added.

[0202] Different N-terminally truncated hST6Gal-I enzyme variants were tested for CMP-dependent sialidase activity: [0203] .DELTA.89 hST6Gal-I (Example 9) [0204] .DELTA.108 hST6Gal-I (Example 9) [0205] .DELTA.57 hST3Gal-I (obtained from R&D Systems)

[0206] Four different experiments were made using .DELTA.89 hST6Gal-I (16.8 .mu.L with 125 .mu.g), .DELTA.108 hST6Gal-I (17.3 .mu.L with 125 .mu.g), .DELTA.57 hST3Gal-I (20.1 .mu.L with 125 .mu.g) and a negative control (no enzyme, 20.1 .mu.L ultrapure water). The enzymes were tested for sialidase activity in the absence and presence of CMP (10-fold excess based on molarity). The concentrations were as shown as follows:

.DELTA.89 hST6Gal-I (16.8 .mu.L with 125 .mu.g): 11.8 .mu.g CMP (c=0.5 mg/mL 23.6 .mu.L) .DELTA.108 hST6Gal-I (17.3 .mu.L with 125 .mu.g): 12.3 .mu.g CMP (c=0.5 mg/mL 24.5 .mu.L) .DELTA.57 hST3Gal-I (20.1 .mu.L with 125 .mu.g): 12.3 .mu.g CMP (c=0.5 mg/mL 24.5 .mu.L) Negative control (no enzyme): 12.3 .mu.g CMP (c=0.5 mg/mL 24.5 .mu.L)

[0207] The samples were incubated at 37.degree. C. in 20 mM sodium citrate, 35 mM Tris pH 6.5. Aliquots were taken as samples after different incubation times, and were analyzed.

[0208] To analyze the degree of sialylation of IgG1 MAB in the samples, 124 .mu.L denaturing buffer (6 M Guanidinium chloride in water) and 30 .mu.L TCEP (=tris(2-carboxyethyl)phosphine; 0.1 mM, diluted in denaturing buffer) were added to 76 .mu.L (corresponding to 250 .mu.g IgG1 MAB) and the sample was incubated at 37.degree. C. for 1 h. After that the sample was buffered in electrospray medium (20% ACN (=acetonitrile), 1% FA (=formamide)) using pre-equilibrated Illustra.TM. Nap5-Columns (GE-Healthcare). Subsequently the sample was analyzed by electrospray ionization mass spectrometry and the content of G2+0SA, G2+1SA and G2+2SA N-glycans was determined. A Synapt G2 HDMS device (Waters UK) was used, the software used was MassLynx V 4.1.

[0209] The results are shown in FIGS. 7-10. In the reaction mixture without CMP no degradation of G2+2SA was observed even after incubation for 46 h (FIG. 7). Whereas in the presence of CMP a degradation of G2+2SA was measured accompanied by an increase of the content of G2+1SA (FIG. 8). Under the conditions described above .DELTA.89 hST6Gal-I showed a CMP-dependent sialidase activity, whereas .DELTA.108 hST6Gal-I (FIG. 9) and .DELTA.57 ST3Gal-I (FIG. 10) did not show any sialidase activity in the presence of CMP. In the latter case this is noted that the enzyme is specific for 2-3 glycosidic bonds

Example 15

[0210] Sialylation of IgG4 MAB Using .DELTA.89 hST6Gal-I in the Presence of Phosphatase Enzymatic Activity

[0211] Suppression of CMP-dependent sialidase activity of .DELTA.89 hST6Gal-I was studied by continuous removal of CMP formed during the reaction.

[0212] In the experiments the enzymes (i) 5'-nucleotidase (EC 3.1.3.5) having a wide specificity for 5'-nucleotides, and (ii) alkaline phosphatase (EC 3.1.3.1) (both provided by commercial suppliers) were used. The particular 5'-nucleotidase used here is also known as ecto-5'-nucleotidase or CD73 (Cluster of Differentiation 73), in humans encoded by the NT5E gene. Both enzymes dephosphorylate CMP, i.e. catalyze hydrolysis of the phosphoester bond in CMP. In the experiments of this Example the enzymes were used to degrade CMP generated by the sialyltransferase reaction from the co-substrate CMP-NANA. In the absence of CMP no intrinsic sialidase activity of .DELTA.89 hST6Gal-I was observed.

[0213] To 1,000 .mu.g IgG4 MAB (182 .mu.L) 500 .mu.g CMP-NANA (3 mg/mL, 166.7 .mu.L), 100 .mu.g .DELTA.89 hST6Gal-I (13.4 .mu.L, 30 .mu.g/300 .mu.g IgG4 MAB) and different amounts of nucleotidase (Nu) and alkaline phosphatase (AP) were added. As Zn.sup.2+ ions are essential for the activity of AP, these were added to a final concentration of 0.1 mM). The buffer used was 20 mM sodium acetate/Tris, pH 6.5.

[0214] Different amounts of the enzymes were added to the reaction mixtures to study the effect of the dephosphorylating enzymes: [0215] 1) 5'-nucleotidase CD73 was used in a concentration of 0.1 .mu.g/.mu.L. To the reactions 0.10 .mu.g, 0.25 .mu.g and 0.50 .mu.g were added. [0216] 2) Alkaline phosphatase (AP) was used in a concentration of 1 .mu.g/.mu.L and 10 .mu.g/.mu.L. To the reactions 1 .mu.g, 5 .mu.g, 10 .mu.g and 100 .mu.g were added.

[0217] After addition of the respective amounts of enzymes the samples were incubated at 37.degree. C. Samples were taken at several time points to control the degree of sialylation. Therefore 110 .mu.L denaturing buffer (6 M Guanidinium chloride) and 30 .mu.L TCEP (0.1 mM, diluted in denaturing buffer) were added to 90 .mu.L of the sample (about 250 .mu.g IgG4 MAB) and the sample was incubated at 37.degree. C. for 1 h. After that the sample was buffered in electrospray medium [20% ACN (=acetonitrile), 1% FA (=formamide)] using pre-equilibrated Illustra.TM. Nap5-Columns (GE-Healthcare). Then the sample was analyzed by electrospray ionization mass spectrometry, and the content of G2+0SA, G2+1SA and G2+2SA N-glycans was determined. A Synapt G2 HDMS device (Waters UK) were used, the software used was MassLynx V 4.1.

[0218] Results for sialylation of IgG4 MAB by .DELTA.89 hST6Gal-I in the absence or presence of 5'-nucleotidase CD73 are depicted in FIGS. 11-14; and results for sialylation of IgG4 MAB by .DELTA.89 hST6Gal-I in the absence or presence of alkaline phosphatase are depicted in FIGS. 15-19. As it turned out, introducing a phosphatase activity capable of hydrolyzing the phosphoester bond in 5'-CMP effectively reduced CMP-mediated sialidase activity and promoted sialyltransferase activity.

Sequence CWU 1

1

71406PRTHomo sapiensMISC_FEATUREhST6-Gal-I WT polypeptide 1Met Ile His Thr Asn Leu Lys Lys Lys Phe Ser Cys Cys Val Leu Val1 5 10 15Phe Leu Leu Phe Ala Val Ile Cys Val Trp Lys Glu Lys Lys Lys Gly 20 25 30Ser Tyr Tyr Asp Ser Phe Lys Leu Gln Thr Lys Glu Phe Gln Val Leu 35 40 45Lys Ser Leu Gly Lys Leu Ala Met Gly Ser Asp Ser Gln Ser Val Ser 50 55 60Ser Ser Ser Thr Gln Asp Pro His Arg Gly Arg Gln Thr Leu Gly Ser65 70 75 80Leu Arg Gly Leu Ala Lys Ala Lys Pro Glu Ala Ser Phe Gln Val Trp 85 90 95Asn Lys Asp Ser Ser Ser Lys Asn Leu Ile Pro Arg Leu Gln Lys Ile 100 105 110Trp Lys Asn Tyr Leu Ser Met Asn Lys Tyr Lys Val Ser Tyr Lys Gly 115 120 125Pro Gly Pro Gly Ile Lys Phe Ser Ala Glu Ala Leu Arg Cys His Leu 130 135 140Arg Asp His Val Asn Val Ser Met Val Glu Val Thr Asp Phe Pro Phe145 150 155 160Asn Thr Ser Glu Trp Glu Gly Tyr Leu Pro Lys Glu Ser Ile Arg Thr 165 170 175Lys Ala Gly Pro Trp Gly Arg Cys Ala Val Val Ser Ser Ala Gly Ser 180 185 190Leu Lys Ser Ser Gln Leu Gly Arg Glu Ile Asp Asp His Asp Ala Val 195 200 205Leu Arg Phe Asn Gly Ala Pro Thr Ala Asn Phe Gln Gln Asp Val Gly 210 215 220Thr Lys Thr Thr Ile Arg Leu Met Asn Ser Gln Leu Val Thr Thr Glu225 230 235 240Lys Arg Phe Leu Lys Asp Ser Leu Tyr Asn Glu Gly Ile Leu Ile Val 245 250 255Trp Asp Pro Ser Val Tyr His Ser Asp Ile Pro Lys Trp Tyr Gln Asn 260 265 270Pro Asp Tyr Asn Phe Phe Asn Asn Tyr Lys Thr Tyr Arg Lys Leu His 275 280 285Pro Asn Gln Pro Phe Tyr Ile Leu Lys Pro Gln Met Pro Trp Glu Leu 290 295 300Trp Asp Ile Leu Gln Glu Ile Ser Pro Glu Glu Ile Gln Pro Asn Pro305 310 315 320Pro Ser Ser Gly Met Leu Gly Ile Ile Ile Met Met Thr Leu Cys Asp 325 330 335Gln Val Asp Ile Tyr Glu Phe Leu Pro Ser Lys Arg Lys Thr Asp Val 340 345 350Cys Tyr Tyr Tyr Gln Lys Phe Phe Asp Ser Ala Cys Thr Met Gly Ala 355 360 365Tyr His Pro Leu Leu Tyr Glu Lys Asn Leu Val Lys His Leu Asn Gln 370 375 380Gly Thr Asp Glu Asp Ile Tyr Leu Leu Gly Lys Ala Thr Leu Pro Gly385 390 395 400Phe Arg Thr Ile His Cys 4052317PRTArtificial Sequencedelta89 truncation variant of hST6Gal-I 2Glu Ala Ser Phe Gln Val Trp Asn Lys Asp Ser Ser Ser Lys Asn Leu1 5 10 15Ile Pro Arg Leu Gln Lys Ile Trp Lys Asn Tyr Leu Ser Met Asn Lys 20 25 30Tyr Lys Val Ser Tyr Lys Gly Pro Gly Pro Gly Ile Lys Phe Ser Ala 35 40 45Glu Ala Leu Arg Cys His Leu Arg Asp His Val Asn Val Ser Met Val 50 55 60Glu Val Thr Asp Phe Pro Phe Asn Thr Ser Glu Trp Glu Gly Tyr Leu65 70 75 80Pro Lys Glu Ser Ile Arg Thr Lys Ala Gly Pro Trp Gly Arg Cys Ala 85 90 95Val Val Ser Ser Ala Gly Ser Leu Lys Ser Ser Gln Leu Gly Arg Glu 100 105 110Ile Asp Asp His Asp Ala Val Leu Arg Phe Asn Gly Ala Pro Thr Ala 115 120 125Asn Phe Gln Gln Asp Val Gly Thr Lys Thr Thr Ile Arg Leu Met Asn 130 135 140Ser Gln Leu Val Thr Thr Glu Lys Arg Phe Leu Lys Asp Ser Leu Tyr145 150 155 160Asn Glu Gly Ile Leu Ile Val Trp Asp Pro Ser Val Tyr His Ser Asp 165 170 175Ile Pro Lys Trp Tyr Gln Asn Pro Asp Tyr Asn Phe Phe Asn Asn Tyr 180 185 190Lys Thr Tyr Arg Lys Leu His Pro Asn Gln Pro Phe Tyr Ile Leu Lys 195 200 205Pro Gln Met Pro Trp Glu Leu Trp Asp Ile Leu Gln Glu Ile Ser Pro 210 215 220Glu Glu Ile Gln Pro Asn Pro Pro Ser Ser Gly Met Leu Gly Ile Ile225 230 235 240Ile Met Met Thr Leu Cys Asp Gln Val Asp Ile Tyr Glu Phe Leu Pro 245 250 255Ser Lys Arg Lys Thr Asp Val Cys Tyr Tyr Tyr Gln Lys Phe Phe Asp 260 265 270Ser Ala Cys Thr Met Gly Ala Tyr His Pro Leu Leu Tyr Glu Lys Asn 275 280 285Leu Val Lys His Leu Asn Gln Gly Thr Asp Glu Asp Ile Tyr Leu Leu 290 295 300Gly Lys Ala Thr Leu Pro Gly Phe Arg Thr Ile His Cys305 310 31531054DNAArtificial SequenceExpression construct Epo-AP-delta89 ST6 (90-406)misc_recomb(1)..(6)Sal-I restriction siteCDS(8)..(1048)nucleic acid sequence encoding a fusion polypeptide of delta89 hST6-Gal-I, N-terminally fused to the Epo leader peptide and containing an "AP" joining sequencemisc_feature(95)..(1048)portion ot the nucleic acid sequence encoding the delta89 hST6-Gal-I portion of the fusion polypeptidemisc_recomb(1049)..(1054)BamH-I restriction site 3gtcgacc atg ggc gtg cac gaa tgt cct gcc tgg ctg tgg ctg ctg ctg 49 Met Gly Val His Glu Cys Pro Ala Trp Leu Trp Leu Leu Leu 1 5 10agc ctg ctg tct ctg cct ctg gga ctg cct gtg ctg ggc gcc cct gaa 97Ser Leu Leu Ser Leu Pro Leu Gly Leu Pro Val Leu Gly Ala Pro Glu15 20 25 30gcc tct ttc cag gtg tgg aac aag gac agc agc tcc aag aac ctg atc 145Ala Ser Phe Gln Val Trp Asn Lys Asp Ser Ser Ser Lys Asn Leu Ile 35 40 45ccc cgg ctg cag aag atc tgg aag aac tac ctg agc atg aac aag tac 193Pro Arg Leu Gln Lys Ile Trp Lys Asn Tyr Leu Ser Met Asn Lys Tyr 50 55 60aag gtg tcc tac aag ggc cct ggc cct ggc atc aag ttt agc gcc gag 241Lys Val Ser Tyr Lys Gly Pro Gly Pro Gly Ile Lys Phe Ser Ala Glu 65 70 75gcc ctg aga tgc cac ctg agg gat cac gtg aac gtg tcc atg gtg gaa 289Ala Leu Arg Cys His Leu Arg Asp His Val Asn Val Ser Met Val Glu 80 85 90gtg acc gac ttc cca ttc aac acc agc gag tgg gag ggc tac ctg ccc 337Val Thr Asp Phe Pro Phe Asn Thr Ser Glu Trp Glu Gly Tyr Leu Pro95 100 105 110aaa gag agc atc cgg acc aaa gcc ggc cct tgg gga aga tgt gcc gtg 385Lys Glu Ser Ile Arg Thr Lys Ala Gly Pro Trp Gly Arg Cys Ala Val 115 120 125gtg tct agc gcc ggc agc ctg aag agt agc cag ctg ggc aga gag atc 433Val Ser Ser Ala Gly Ser Leu Lys Ser Ser Gln Leu Gly Arg Glu Ile 130 135 140gac gac cac gac gcc gtg ctg cgg ttc aat ggc gct ccc acc gcc aac 481Asp Asp His Asp Ala Val Leu Arg Phe Asn Gly Ala Pro Thr Ala Asn 145 150 155ttc cag cag gac gtg ggc acc aag acc acc atc cgg ctg atg aac tcc 529Phe Gln Gln Asp Val Gly Thr Lys Thr Thr Ile Arg Leu Met Asn Ser 160 165 170cag ctc gtg aca acc gag aag cgg ttc ctg aag gac agc ctg tac aac 577Gln Leu Val Thr Thr Glu Lys Arg Phe Leu Lys Asp Ser Leu Tyr Asn175 180 185 190gag ggc atc ctg atc gtg tgg gac ccc agc gtg tac cac agc gac atc 625Glu Gly Ile Leu Ile Val Trp Asp Pro Ser Val Tyr His Ser Asp Ile 195 200 205ccc aag tgg tat cag aac ccc gac tac aac ttc ttc aac aac tac aag 673Pro Lys Trp Tyr Gln Asn Pro Asp Tyr Asn Phe Phe Asn Asn Tyr Lys 210 215 220acc tac cgg aag ctg cac ccc aac cag ccc ttc tac atc ctg aag ccc 721Thr Tyr Arg Lys Leu His Pro Asn Gln Pro Phe Tyr Ile Leu Lys Pro 225 230 235cag atg ccc tgg gag ctg tgg gac att ctg cag gaa atc agc ccc gaa 769Gln Met Pro Trp Glu Leu Trp Asp Ile Leu Gln Glu Ile Ser Pro Glu 240 245 250gag atc cag ccc aac ccc cct agc tct ggc atg ctg ggc atc att atc 817Glu Ile Gln Pro Asn Pro Pro Ser Ser Gly Met Leu Gly Ile Ile Ile255 260 265 270atg atg acc ctg tgc gac cag gtg gac atc tac gag ttt ctg ccc tcc 865Met Met Thr Leu Cys Asp Gln Val Asp Ile Tyr Glu Phe Leu Pro Ser 275 280 285aag aga aag acc gac gtg tgc tac tac tac cag aag ttc ttc gac agc 913Lys Arg Lys Thr Asp Val Cys Tyr Tyr Tyr Gln Lys Phe Phe Asp Ser 290 295 300gcc tgc acc atg gga gcc tac cac cct ctg ctg tac gag aag aac ctc 961Ala Cys Thr Met Gly Ala Tyr His Pro Leu Leu Tyr Glu Lys Asn Leu 305 310 315gtg aag cac ctg aac cag ggc acc gac gag gat atc tac ctg ctg ggc 1009Val Lys His Leu Asn Gln Gly Thr Asp Glu Asp Ile Tyr Leu Leu Gly 320 325 330aag gcc acc ctg ccc ggc ttc aga acc atc cac tgc tga ggatcc 1054Lys Ala Thr Leu Pro Gly Phe Arg Thr Ile His Cys335 340 3454346PRTArtificial SequenceSynthetic Construct 4Met Gly Val His Glu Cys Pro Ala Trp Leu Trp Leu Leu Leu Ser Leu1 5 10 15Leu Ser Leu Pro Leu Gly Leu Pro Val Leu Gly Ala Pro Glu Ala Ser 20 25 30Phe Gln Val Trp Asn Lys Asp Ser Ser Ser Lys Asn Leu Ile Pro Arg 35 40 45Leu Gln Lys Ile Trp Lys Asn Tyr Leu Ser Met Asn Lys Tyr Lys Val 50 55 60Ser Tyr Lys Gly Pro Gly Pro Gly Ile Lys Phe Ser Ala Glu Ala Leu65 70 75 80Arg Cys His Leu Arg Asp His Val Asn Val Ser Met Val Glu Val Thr 85 90 95Asp Phe Pro Phe Asn Thr Ser Glu Trp Glu Gly Tyr Leu Pro Lys Glu 100 105 110Ser Ile Arg Thr Lys Ala Gly Pro Trp Gly Arg Cys Ala Val Val Ser 115 120 125Ser Ala Gly Ser Leu Lys Ser Ser Gln Leu Gly Arg Glu Ile Asp Asp 130 135 140His Asp Ala Val Leu Arg Phe Asn Gly Ala Pro Thr Ala Asn Phe Gln145 150 155 160Gln Asp Val Gly Thr Lys Thr Thr Ile Arg Leu Met Asn Ser Gln Leu 165 170 175Val Thr Thr Glu Lys Arg Phe Leu Lys Asp Ser Leu Tyr Asn Glu Gly 180 185 190Ile Leu Ile Val Trp Asp Pro Ser Val Tyr His Ser Asp Ile Pro Lys 195 200 205Trp Tyr Gln Asn Pro Asp Tyr Asn Phe Phe Asn Asn Tyr Lys Thr Tyr 210 215 220Arg Lys Leu His Pro Asn Gln Pro Phe Tyr Ile Leu Lys Pro Gln Met225 230 235 240Pro Trp Glu Leu Trp Asp Ile Leu Gln Glu Ile Ser Pro Glu Glu Ile 245 250 255Gln Pro Asn Pro Pro Ser Ser Gly Met Leu Gly Ile Ile Ile Met Met 260 265 270Thr Leu Cys Asp Gln Val Asp Ile Tyr Glu Phe Leu Pro Ser Lys Arg 275 280 285Lys Thr Asp Val Cys Tyr Tyr Tyr Gln Lys Phe Phe Asp Ser Ala Cys 290 295 300Thr Met Gly Ala Tyr His Pro Leu Leu Tyr Glu Lys Asn Leu Val Lys305 310 315 320His Leu Asn Gln Gly Thr Asp Glu Asp Ile Tyr Leu Leu Gly Lys Ala 325 330 335Thr Leu Pro Gly Phe Arg Thr Ile His Cys 340 3455298PRTArtificial Sequencedelta108 truncation variant of hST6Gal-I 5Leu Gln Lys Ile Trp Lys Asn Tyr Leu Ser Met Asn Lys Tyr Lys Val1 5 10 15Ser Tyr Lys Gly Pro Gly Pro Gly Ile Lys Phe Ser Ala Glu Ala Leu 20 25 30Arg Cys His Leu Arg Asp His Val Asn Val Ser Met Val Glu Val Thr 35 40 45Asp Phe Pro Phe Asn Thr Ser Glu Trp Glu Gly Tyr Leu Pro Lys Glu 50 55 60Ser Ile Arg Thr Lys Ala Gly Pro Trp Gly Arg Cys Ala Val Val Ser65 70 75 80Ser Ala Gly Ser Leu Lys Ser Ser Gln Leu Gly Arg Glu Ile Asp Asp 85 90 95His Asp Ala Val Leu Arg Phe Asn Gly Ala Pro Thr Ala Asn Phe Gln 100 105 110Gln Asp Val Gly Thr Lys Thr Thr Ile Arg Leu Met Asn Ser Gln Leu 115 120 125Val Thr Thr Glu Lys Arg Phe Leu Lys Asp Ser Leu Tyr Asn Glu Gly 130 135 140Ile Leu Ile Val Trp Asp Pro Ser Val Tyr His Ser Asp Ile Pro Lys145 150 155 160Trp Tyr Gln Asn Pro Asp Tyr Asn Phe Phe Asn Asn Tyr Lys Thr Tyr 165 170 175Arg Lys Leu His Pro Asn Gln Pro Phe Tyr Ile Leu Lys Pro Gln Met 180 185 190Pro Trp Glu Leu Trp Asp Ile Leu Gln Glu Ile Ser Pro Glu Glu Ile 195 200 205Gln Pro Asn Pro Pro Ser Ser Gly Met Leu Gly Ile Ile Ile Met Met 210 215 220Thr Leu Cys Asp Gln Val Asp Ile Tyr Glu Phe Leu Pro Ser Lys Arg225 230 235 240Lys Thr Asp Val Cys Tyr Tyr Tyr Gln Lys Phe Phe Asp Ser Ala Cys 245 250 255Thr Met Gly Ala Tyr His Pro Leu Leu Tyr Glu Lys Asn Leu Val Lys 260 265 270His Leu Asn Gln Gly Thr Asp Glu Asp Ile Tyr Leu Leu Gly Lys Ala 275 280 285Thr Leu Pro Gly Phe Arg Thr Ile His Cys 290 29561003DNAArtificial SequenceExpression construct Epo-AP-delta108 ST6 (109-406)CDS(8)..(997)open reading frame 6gtcgacc atg ggc gtg cac gaa tgt cct gcc tgg ctg tgg ctg ctg ctg 49 Met Gly Val His Glu Cys Pro Ala Trp Leu Trp Leu Leu Leu 1 5 10agc ctg ctg tct ctg cct ctg gga ctg cct gtg ctg ggc gcc cct cct 97Ser Leu Leu Ser Leu Pro Leu Gly Leu Pro Val Leu Gly Ala Pro Pro15 20 25 30aga ctg cag aag atc tgg aag aac tac ctg agc atg aac aag tac aag 145Arg Leu Gln Lys Ile Trp Lys Asn Tyr Leu Ser Met Asn Lys Tyr Lys 35 40 45gtg tcc tac aag ggc cct ggc cct ggc atc aag ttt agc gcc gag gcc 193Val Ser Tyr Lys Gly Pro Gly Pro Gly Ile Lys Phe Ser Ala Glu Ala 50 55 60ctg aga tgc cac ctg agg gat cac gtg aac gtg tcc atg gtg gaa gtg 241Leu Arg Cys His Leu Arg Asp His Val Asn Val Ser Met Val Glu Val 65 70 75acc gac ttc cca ttc aac acc agc gag tgg gag ggc tac ctg ccc aaa 289Thr Asp Phe Pro Phe Asn Thr Ser Glu Trp Glu Gly Tyr Leu Pro Lys 80 85 90gag agc atc cgg acc aaa gcc ggc cct tgg gga aga tgt gcc gtg gtg 337Glu Ser Ile Arg Thr Lys Ala Gly Pro Trp Gly Arg Cys Ala Val Val95 100 105 110tct agc gcc ggc agc ctg aag agt agc cag ctg ggc aga gag atc gac 385Ser Ser Ala Gly Ser Leu Lys Ser Ser Gln Leu Gly Arg Glu Ile Asp 115 120 125gac cac gac gcc gtg ctg cgg ttc aat ggc gct ccc acc gcc aac ttc 433Asp His Asp Ala Val Leu Arg Phe Asn Gly Ala Pro Thr Ala Asn Phe 130 135 140cag cag gac gtg ggc acc aag acc acc atc cgg ctg atg aac tcc cag 481Gln Gln Asp Val Gly Thr Lys Thr Thr Ile Arg Leu Met Asn Ser Gln 145 150 155ctc gtg aca acc gag aag cgg ttc ctg aag gac agc ctg tac aac gag 529Leu Val Thr Thr Glu Lys Arg Phe Leu Lys Asp Ser Leu Tyr Asn Glu 160 165 170ggc atc ctg atc gtg tgg gac ccc agc gtg tac cac agc gac atc ccc 577Gly Ile Leu Ile Val Trp Asp Pro Ser Val Tyr His Ser Asp Ile Pro175 180 185 190aag tgg tat cag aac ccc gac tac aac ttc ttc aac aac tac aag acc 625Lys Trp Tyr Gln Asn Pro Asp Tyr Asn Phe Phe Asn Asn Tyr Lys Thr 195 200 205tac cgg aag ctg cac ccc aac cag ccc ttc tac atc ctg aag ccc cag 673Tyr Arg Lys Leu His Pro Asn Gln Pro Phe Tyr Ile Leu Lys Pro Gln 210 215 220atg ccc tgg gag ctg tgg gac att ctg cag gaa atc agc ccc gaa gag 721Met Pro Trp Glu Leu Trp Asp Ile Leu Gln Glu Ile Ser Pro Glu Glu 225 230 235atc cag ccc aac ccc cct agc tct ggc atg ctg ggc atc att atc atg 769Ile Gln Pro Asn Pro Pro Ser Ser Gly Met Leu Gly Ile Ile Ile Met 240 245 250atg acc ctg tgc gac cag gtg gac atc tac gag ttt ctg ccc tcc aag 817Met Thr Leu Cys Asp Gln Val Asp Ile Tyr Glu Phe Leu Pro Ser Lys255

260 265 270aga aag acc gac gtg tgc tac tac tac cag aag ttc ttc gac agc gcc 865Arg Lys Thr Asp Val Cys Tyr Tyr Tyr Gln Lys Phe Phe Asp Ser Ala 275 280 285tgc acc atg gga gcc tac cac cct ctg ctg tac gag aag aac ctc gtg 913Cys Thr Met Gly Ala Tyr His Pro Leu Leu Tyr Glu Lys Asn Leu Val 290 295 300aag cac ctg aac cag ggc acc gac gag gat atc tac ctg ctg ggc aag 961Lys His Leu Asn Gln Gly Thr Asp Glu Asp Ile Tyr Leu Leu Gly Lys 305 310 315gcc acc ctg ccc ggc ttc aga acc atc cac tgc tga ggatcc 1003Ala Thr Leu Pro Gly Phe Arg Thr Ile His Cys 320 3257329PRTArtificial SequenceSynthetic Construct 7Met Gly Val His Glu Cys Pro Ala Trp Leu Trp Leu Leu Leu Ser Leu1 5 10 15Leu Ser Leu Pro Leu Gly Leu Pro Val Leu Gly Ala Pro Pro Arg Leu 20 25 30Gln Lys Ile Trp Lys Asn Tyr Leu Ser Met Asn Lys Tyr Lys Val Ser 35 40 45Tyr Lys Gly Pro Gly Pro Gly Ile Lys Phe Ser Ala Glu Ala Leu Arg 50 55 60Cys His Leu Arg Asp His Val Asn Val Ser Met Val Glu Val Thr Asp65 70 75 80Phe Pro Phe Asn Thr Ser Glu Trp Glu Gly Tyr Leu Pro Lys Glu Ser 85 90 95Ile Arg Thr Lys Ala Gly Pro Trp Gly Arg Cys Ala Val Val Ser Ser 100 105 110Ala Gly Ser Leu Lys Ser Ser Gln Leu Gly Arg Glu Ile Asp Asp His 115 120 125Asp Ala Val Leu Arg Phe Asn Gly Ala Pro Thr Ala Asn Phe Gln Gln 130 135 140Asp Val Gly Thr Lys Thr Thr Ile Arg Leu Met Asn Ser Gln Leu Val145 150 155 160Thr Thr Glu Lys Arg Phe Leu Lys Asp Ser Leu Tyr Asn Glu Gly Ile 165 170 175Leu Ile Val Trp Asp Pro Ser Val Tyr His Ser Asp Ile Pro Lys Trp 180 185 190Tyr Gln Asn Pro Asp Tyr Asn Phe Phe Asn Asn Tyr Lys Thr Tyr Arg 195 200 205Lys Leu His Pro Asn Gln Pro Phe Tyr Ile Leu Lys Pro Gln Met Pro 210 215 220Trp Glu Leu Trp Asp Ile Leu Gln Glu Ile Ser Pro Glu Glu Ile Gln225 230 235 240Pro Asn Pro Pro Ser Ser Gly Met Leu Gly Ile Ile Ile Met Met Thr 245 250 255Leu Cys Asp Gln Val Asp Ile Tyr Glu Phe Leu Pro Ser Lys Arg Lys 260 265 270Thr Asp Val Cys Tyr Tyr Tyr Gln Lys Phe Phe Asp Ser Ala Cys Thr 275 280 285Met Gly Ala Tyr His Pro Leu Leu Tyr Glu Lys Asn Leu Val Lys His 290 295 300Leu Asn Gln Gly Thr Asp Glu Asp Ile Tyr Leu Leu Gly Lys Ala Thr305 310 315 320Leu Pro Gly Phe Arg Thr Ile His Cys 325

* * * * *

References


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed